1
|
Pimenta JC, Beltrami VA, Oliveira BDS, Queiroz-Junior CM, Barsalini J, Teixeira DC, de Souza-Costa LP, Lima ALD, Machado CA, Parreira BZSG, Santos FRDS, Costa PAC, Lacerda LDSB, Gonçalves MR, Chaves IDM, Couto MGG, Costa VRDM, Nóbrega NRC, Silva BL, Fonseca T, Resende F, Wnuk NT, Marim FM, Rocha FEO, Umezu HL, Campolina-Silva G, Andrade ACDSP, de Aguiar RS, Costa GMJ, Guimarães PPG, Silva GSF, Rachid MA, Vieira LB, Pinho V, Teixeira AL, Teixeira MM, Miranda AS, Costa VV. Neuropsychiatric sequelae in an experimental model of post-COVID syndrome in mice. Brain Behav Immun 2025; 128:16-36. [PMID: 40120834 DOI: 10.1016/j.bbi.2025.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/17/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
The global impact of the COVID-19 pandemic has been unprecedented, and presently, the world is facing a new challenge known as post-COVID syndrome (PCS). Current estimates suggest that more than 100 million people are grappling with PCS, encompassing several manifestations, including pulmonary, musculoskeletal, metabolic, and neuropsychiatric sequelae (cognitive and behavioral). The mechanisms underlying PCS remain unclear. The present study aimed to: (i) comprehensively characterize the acute effects of pulmonary inoculation of the betacoronavirus MHV-A59 in immunocompetent mice at clinical, cellular, and molecular levels; (ii) examine potential acute and long-term pulmonary, musculoskeletal, and neuropsychiatric sequelae induced by the betacoronavirus MHV-A59; and to (iii) assess sex-specific differences. Male and female C57Bl/6 mice were initially inoculated with varying viral titers (3x103 to 3x105 PFU/30 μL) of the betacoronavirus MHV-A59 via the intranasal route to define the highest inoculum capable of inducing disease without causing mortality. Further experiments were conducted with the 3x104 PFU inoculum. Mice exhibited an altered neutrophil/lymphocyte ratio in the blood in the 2nd and 5th day post-infection (dpi). Marked lung lesions were characterized by hyperplasia of the alveolar walls, infiltration of polymorphonuclear leukocytes (PMN) and mononuclear leukocytes, hemorrhage, increased concentrations of CCL2, CCL3, CCL5, and CXCL1 chemokines, as well as high viral titers until the 5th dpi. While these lung inflammatory signs resolved, other manifestations were observed up to the 60 dpi, including mild brain lesions with gliosis and hyperemic blood vessels, neuromuscular dysfunctions, anhedonic-like behavior, deficits in spatial working memory, and short-term aversive memory. These musculoskeletal and neuropsychiatric complications were exclusive to female mice and prevented after ovariectomy. In summary, our study describes for the first time a novel sex-dependent model of PCS focused on neuropsychiatric and musculoskeletal disorders. This model provides a unique platform for future investigations regarding the effects of acute therapeutic interventions on the long-term sequelae unleashed by betacoronavirus infection.
Collapse
Affiliation(s)
- Jordane Clarisse Pimenta
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vinícius Amorim Beltrami
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Bruna da Silva Oliveira
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Celso Martins Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jéssica Barsalini
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Danielle Cunha Teixeira
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luiz Pedro de Souza-Costa
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Anna Luiza Diniz Lima
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Caroline Amaral Machado
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Felipe Rocha da Silva Santos
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro Augusto Carvalho Costa
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Matheus Rodrigues Gonçalves
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ian de Meira Chaves
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Manoela Gonzaga Gontijo Couto
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Victor Rodrigues de Melo Costa
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Bárbara Luísa Silva
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Talita Fonseca
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Filipe Resende
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Natália Teixeira Wnuk
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fernanda Martins Marim
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Felipe Emanuel Oliveira Rocha
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Hanna L Umezu
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gabriel Campolina-Silva
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Department of Obstetrics, Gynecology and Reproduction, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Ana Cláudia Dos Santos Pereira Andrade
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Department of Microbiology and Immunology, Université Laval, Quebec, Canada
| | - Renato Santana de Aguiar
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Guilherme Mattos Jardim Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro Pires Goulart Guimarães
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Glauber Santos Ferreira Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Milene Alvarenga Rachid
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luciene Bruno Vieira
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vanessa Pinho
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Antônio Lúcio Teixeira
- Biggs Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Aline Silva Miranda
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Vivian Vasconcelos Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
2
|
Hashemipour S, Kalantarian SS, Panahi H, Kelishomi SE, Ghasemi A, Chopani SM, Kolaji S, Badri M, Ghobadi A, Khairkhahan SMRH, Lalooha F, Movahed F, Abbasi M, Elmizadeh K. The association of inflammatory markers in early pregnancy with the development of gestational diabetes: Qazvin maternal and neonatal metabolic study (QMNS). BMC Pregnancy Childbirth 2025; 25:135. [PMID: 39934746 PMCID: PMC11816776 DOI: 10.1186/s12884-025-07267-y] [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: 02/20/2024] [Accepted: 01/31/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND This study investigates the association of blood cell components and blood cell-derived inflammatory indices in early pregnancy with the development of gestational diabetes mellitus (GDM). METHODS This research is part of the Qazvin maternal and neonatal metabolic study (QMNMS) conducted in Iran from 2018 to 2021. Pregnant women with gestational age ≤ 14 weeks were enrolled in the study. The association of blood cells and inflammatory indices, including neutrophil-to-lymphocyte ratio (NLR), systemic inflammatory response index (SIRI), systemic immune inflammation index (SII), and aggregate systemic inflammatory response index (AISI), in early pregnancy with subsequent GDM development was examined using multivariate logistic regression. This analysis was adjusted for age, pre-pregnancy body mass index (BMI), weight gain, and GDM history in previous pregnancies (Model 1), as well as for these factors in addition to the homeostatic model assessment for insulin resistance (HOMA-IR) (Model 2). The correlation of blood cells and inflammatory indices with insulin resistance was assessed through Spearman partial correlation, adjusted for the same risk factors. RESULTS The final analysis included 612 participants, among whom GDM developed in 96 participants (15.7%). Neutrophil, platelet, and lymphocyte counts showed significant correlations with HOMA-IR (r = 0.14, r = 0.22, and r = 0.17, respectively; P < 0.01 for all). In univariate analysis, the highest quartile of neutrophil count was associated with a 5.9 times higher risk of GDM development (95% CI 2.6-13.2, P < 0.001). In multivariate logistic regression, neutrophil count quartiles remained significant predictors of GDM development, with relative risks of 3.7, 4.4, and 8.2 for the 2nd, 3rd, and 4th neutrophil quartiles compared to the 1st quartile (P < 0.001). While platelet count was initially associated with GDM development (RR = 2.6, 95% CI 1.3-5.1, P = 0.028), this association was no longer significant after adjusting for HOMA-IR. Neither lymphocyte nor monocyte counts were linked to GDM development. Additionally, inflammatory indices, such as NLR, SIRI, SII, and AISI, did not provide additional predictive value for GDM development. CONCLUSION Neutrophil count is an independent predictor of GDM development, and its role in GDM development is not influenced by early pregnancy insulin resistance. Moreover, novel inflammatory indices offer no additional predictive benefit for GDM.
Collapse
Affiliation(s)
- Sima Hashemipour
- Non-Communicable Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Seyedeh Sareh Kalantarian
- Non-Communicable Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Hamidreza Panahi
- Non-Communicable Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sara Esmaeili Kelishomi
- Non-Communicable Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Amirabbas Ghasemi
- Non-Communicable Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sarah Mirzaeei Chopani
- Non-Communicable Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sepideh Kolaji
- Non-Communicable Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Milad Badri
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Arefeh Ghobadi
- Non-Communicable Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | | | - Fatemeh Lalooha
- Non-Communicable Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Farideh Movahed
- Non-Communicable Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mahnaz Abbasi
- Non-Communicable Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Khadijeh Elmizadeh
- Non-Communicable Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran.
| |
Collapse
|
3
|
Dai SZ, Wu RH, Chen H, Chen MH, Xie W, Zheng WP, Tan GH, Huang FY. Progesterone suppresses rhinovirus-induced airway inflammation by inhibiting neutrophil infiltration and extracellular traps formation. Int Immunopharmacol 2025; 144:113714. [PMID: 39626540 DOI: 10.1016/j.intimp.2024.113714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/13/2024] [Accepted: 11/23/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND The process of NETosis is observed in a range of inflammatory conditions. Progesterone (P4) has been shown to alleviate inflammation caused by viral infections such as influenza and SARS-CoV-2. However, the precise molecular mechanisms responsible for this effect are not yet fully understood. Therefore, the present investigation aims to explore whether P4 can exert its anti-inflammatory properties by inhibiting NETosis and the related molecular pathways. METHODS Airway inflammation caused by rhinovirus serotype-1b (RV-1b) was induced in male BALB/c mice. The inflammation was assessed through histological examination and calculation of inflammatory cells present in the bronchoalveolar lavage fluid. Flow cytometry was used to analyze the inflammatory cells and NETotic neutrophils. Western blotting analysis was conducted to detect proteins associated with NETosis, inflammasome activation, and signaling. Furthermore, confocal microscopy was utilized to observe neutrophil extracellular trap (NET) structures in vivo tissues and in vitro neutrophils, neutrophil infiltration, and inflammasome formation. RESULTS The administration of P4 proved to be an effective treatment for reducing airway inflammation and the production of NETs caused by RV-1b infection. The infection triggered the activation of NLRP3 inflammasomes in neutrophils, which led to the maturation of IL-1β and subsequent activation of both the NF-κB and p38 signaling pathways. The activation of NF-κB signaling resulted in the secretion of downstream chemokines CCL3 and IL-6, which led to an increase in neutrophil infiltration into the lung airways. Moreover, the activation of p38 signaling led to the generation of reactive oxygen species, resulting in NETosis. However, the administration of P4 inhibited the activation of the NLRP3 inflammasome, which subsequently led to the deactivation of both the IL-1β-NF-κB and IL-1β-p38 axes. As a result, there was a reduction in neutrophil infiltration and NETosis. Furthermore, TGF-β-activated kinase 1 (TAK1) was identified as an intermediary enzyme. P4 inhibits both the NF-κB and IL-1β-p38 pathways by suppressing the activity of TAK1. CONCLUSION The capacity of P4 to mitigate rhinovirus-induced airway inflammation is attributed to its ability to impede the infiltration of neutrophils and NETosis. As inflammation mediated by NETosis is widespread in diverse disorders, our findings propose that P4 could potentially function as a universal therapeutic agent in the management of such ailments.
Collapse
Affiliation(s)
- Shu-Zhen Dai
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China; Hainan Academy of Medical Sciences, Hainan Medical University, Hainan 571199, China
| | - Ri-Hong Wu
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Hengyu Chen
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Ming-Hui Chen
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Weijing Xie
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Wu-Ping Zheng
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Guang-Hong Tan
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China.
| | - Feng-Ying Huang
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China.
| |
Collapse
|
4
|
Calo G, Merech F, Sabbione F, Hauk V, Lara B, Doga L, D'eramo L, Squassi A, Ramhorst R, Trevani A, Vota D, Leirós CP. Pregnancy Entails a Metabolic Rewiring of Maternal Circulating Neutrophils. J Cell Physiol 2025; 240:e31502. [PMID: 39676603 DOI: 10.1002/jcp.31502] [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: 08/27/2024] [Revised: 10/29/2024] [Accepted: 11/22/2024] [Indexed: 12/17/2024]
Abstract
Immunometabolism is an emerging growing field that focuses on the role of cellular metabolism in the regulation of immune cell function and fate. Thus, proliferation, differentiation, activation, and function of immune cell populations are modulated by reprogramming their fueling and metabolic pathways. Pregnancy entails a fine immune and metabolic regulation of the maternal-fetal interaction to assist the energetic demands of the fetus where trophoblast cells have a central role. Maternal neutrophil functional shaping by trophoblast cells has been proposed though their metabolic conditioning during pregnancy has not been studied yet. Here, we explored the effects of trophoblast-derived factors on the metabolic rewiring of neutrophils from nonpregnant women and its impact on central functions like reactive oxygen species (ROS) production, neutrophil extracellular trap (NET) release, and migration. In parallel, the immunometabolic status and function of neutrophils isolated from pregnant women (16-20 weeks) was compared with nonpregnant age-matched control samples. Trophoblast-derived factors induced glucose uptake and lipid droplet accumulation without activating ROS production or NET release. Conditioned media from trophoblast cells also inhibited PMA-induced NETosis partly by impairing glucose uptake in neutrophils. In turn, neutrophils from pregnant women had increased basal ROS production, lipid accumulation, and glucose uptake compared to neutrophils from nonpregnant women, accompanied by a higher release of PMA-induced NETs. Interestingly, PMA-induced NETs was blocked by a fatty acid oxidation inhibitor in neutrophils from pregnant women indicating the contribution of fatty acid metabolism to neutrophil activity during pregnancy. Results are consistent with immunometabolic mechanisms underlying the functional shaping of neutrophils during pregnancy and point out the contribution of trophoblast-derived factors to their metabolic profiling. These findings provide novel immunometabolic clues to understand immune homeostasis maintenance during pregnancy and raise the clinical potential of monitoring neutrophil metabolism during normal and complicated pregnancies.
Collapse
Affiliation(s)
- Guillermina Calo
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Fátima Merech
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Florencia Sabbione
- Instituto de medicina experimental (IMEX)-CONICET/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Vanesa Hauk
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Brenda Lara
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Luciana Doga
- Universidad de Buenos Aires-Facultad de Odontología, Cátedra de Odontología Preventiva y Comunitaria, Buenos Aires, Argentina
| | - Luciana D'eramo
- Universidad de Buenos Aires-Facultad de Odontología, Cátedra de Odontología Preventiva y Comunitaria, Buenos Aires, Argentina
| | - Aldo Squassi
- Universidad de Buenos Aires-Facultad de Odontología, Cátedra de Odontología Preventiva y Comunitaria, Buenos Aires, Argentina
| | - Rosanna Ramhorst
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Analía Trevani
- Instituto de medicina experimental (IMEX)-CONICET/Academia Nacional de Medicina de Buenos Aires, Buenos Aires, Argentina
| | - Daiana Vota
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Claudia Pérez Leirós
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
5
|
Yang Y, Ru H, Zhang S, Wu C, Dong J, Wang X, Qie Y, Zhang H, Zhang P, Ma J, Du L. The Effect of Granulocyte Colony-Stimulating Factor on Endometrial Receptivity of Implantation Failure Mouse. Reprod Sci 2025; 32:200-217. [PMID: 38600416 DOI: 10.1007/s43032-024-01527-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 03/19/2024] [Indexed: 04/12/2024]
Abstract
The purpose of this study was to investigate the effect of G-CSF on the endometrial receptivity of implantation failure mice. Sixty female mice were treated mifepristone to establish an implant failure model. The treatment groups received different doses of G-CSF. Endometrial tissue and serum were collected on day 5 after mating. The abundance of pinopodes on the endometrium was observed by scanning electron microscopy. The expressions of LPAR3, COX2, and HOXA10 were detected by RT-qPCR and Western blotting. Serum levels of E2, P, VEGF, LIF, TNF-α and IL-10 were measured by ELISA. The expressions of VEGF, CD34, CD57, TNF-α, and IL-10 were assessed by immunohistochemistry. Immunofluorescence analysis was performed to determine the number of CD57, Treg, and Th17 cells. G-CSF increased implantation and pregnancy rates of mifepristone-induced implantation failure mice, with the most significant effect seen at the intermediate dose. G-CSF increased the serum levels of E2 and P, the abundance of endometrial pinopodes, and the level of LIF in the endometrium. It also promoted the expression of VEGF, HOXA10, LPAR3, and COX2. Moreover, G-CSF reduced the level of CD57 cells and the ratio of Th17/Treg cells in endometrium. G-CSF reduced the inflammatory factor TNF-α, but IL-10 did not change significantly. G-CSF can enhance embryo implantation rate and pregnancy rate and improve endometrial receptivity by attenuating degeneration of pinopodes, upregulating estrogen and progesterone, facilitating angiogenesis, maintaining immune cell homeostasis, and reducing the production of inflammatory cytokines in implantation failure mouse.
Collapse
Affiliation(s)
- Yang Yang
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Shijiazhuang, 050071, China
| | - Huibo Ru
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Shijiazhuang, 050071, China
| | - Suzhi Zhang
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Shijiazhuang, 050071, China
| | - Chun Wu
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Shijiazhuang, 050071, China
| | - Jiuhua Dong
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Shijiazhuang, 050071, China
| | - Xiu Wang
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Shijiazhuang, 050071, China
| | - Yaxi Qie
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Shijiazhuang, 050071, China
| | - Hongxia Zhang
- NCPC Genetech Biotechnology Co., Ltd., Shijiazhuang, 050071, China
| | - Peng Zhang
- NCPC Genetech Biotechnology Co., Ltd., Shijiazhuang, 050071, China
| | - Jing Ma
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Shijiazhuang, 050071, China
| | - Lirong Du
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Shijiazhuang, 050071, China.
| |
Collapse
|
6
|
Bun M, Kawano M, Yamamoto G, Sakata M, Shimura K, Toda A, Nakamura K, Kinose Y, Kodama M, Hashimoto K, Kobayashi E, Sawada K, Kimura T. G-CSF induces neutrophil extracellular traps formation and promotes ovarian cancer peritoneal dissemination. J Leukoc Biol 2024; 116:1157-1168. [PMID: 39082070 DOI: 10.1093/jleuko/qiae166] [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: 01/12/2024] [Revised: 06/16/2024] [Accepted: 07/30/2024] [Indexed: 11/05/2024] Open
Abstract
Epithelial ovarian cancer is characterized by aggressive peritoneal dissemination. Neutrophils are mobilized to peritoneal cavity in some patients with ovarian cancer dissemination; however, its pathological significance remains unknown. This study aimed to investigate the role of neutrophil extracellular traps (NETs) in ovarian cancer dissemination. We conducted a retrospective analysis of clinical data and samples from 340 patients with ovarian cancer who underwent primary surgery between 2007 and 2016 at the Osaka University Hospital. In vitro, NETs formation was induced by stimulating human peripheral neutrophils. The human ovarian cancer cell line, OVCAR8, was cocultured with NETs. For an ovarian cancer dissemination mouse model, we performed an intraperitoneal injection of OVCAR8 cells into nude mice. The association between NETs and peritoneal dissemination was explored, and model mice were treated with the PAD4 inhibitor GSK484 to assess antitumor efficacy. Neutrophilia (neutrophil count >7000/mm3) correlated with shorter survival, advanced peritoneal dissemination, elevated granulocyte colony-stimulating factor (G-CSF) levels, increased neutrophil count in ascites, and augmented NETs foci in peritoneal dissemination sites. In vitro assays revealed that G-CSF stimulated neutrophils to form NETs, promoting cancer cell adhesion. In vivo investigations revealed that G-CSF-producing tumor-bearing mice had accelerated peritoneal dissemination and poor prognosis. NETs formation was pathologically observed at the peritoneal dissemination sites. Inhibition of NETs formation by GSK484 significantly delayed peritoneal dissemination in vivo. In conclusion, G-CSF was associated with intra-abdominal NETs formation and increased peritoneal dissemination. NETs represent potential therapeutic targets for ovarian cancer, particularly in patients with neutrophilia.
Collapse
Affiliation(s)
- Michiko Bun
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Mahiru Kawano
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Gaku Yamamoto
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Mina Sakata
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kotaro Shimura
- Department of Obstetrics and Gynecology, Osaka Rosai Hospital, 1179-3, Nakasone, Kita-ku, Sakai, Osaka 591-8025, Japan
| | - Aska Toda
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Koji Nakamura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yasuto Kinose
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Michiko Kodama
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kae Hashimoto
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Eiji Kobayashi
- Department of Obstetrics and Gynecology, Faculty of Medicine, Oita University, Idaigaoka 1-1, Hasama-machi, Yufu, Oita 879-5593, Japan
| | - Kenjiro Sawada
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
7
|
Meier A, Sakoulas G, Nizet V, Ulloa ER. Neutrophil Extracellular Traps: An Emerging Therapeutic Target to Improve Infectious Disease Outcomes. J Infect Dis 2024; 230:514-521. [PMID: 38728418 PMCID: PMC11326844 DOI: 10.1093/infdis/jiae252] [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: 01/29/2024] [Revised: 04/25/2024] [Accepted: 05/09/2024] [Indexed: 05/12/2024] Open
Abstract
Neutrophils possess a diverse repertoire of pathogen clearance mechanisms, one of which is the formation of neutrophil extracellular traps (NETs). NETs are complexes of histone proteins and DNA coated with proteolytic enzymes that are released extracellularly to entrap pathogens and aid in their clearance, in a process known as NETosis. Intravascular NETosis may drive a massive inflammatory response that has been shown to contribute to morbidity and mortality in many infectious diseases, including malaria, dengue fever, influenza, bacterial sepsis, and severe acute respiratory syndrome coronavirus 2 infection. In this review we seek to (1) summarize the current understanding of NETs, (2) discuss infectious diseases in which NET formation contributes to morbidity and mortality, and (3) explore potential adjunctive therapeutics that may be considered for future study in treating severe infections driven by NET pathophysiology. This includes drugs specifically targeting NET inhibition and US Food and Drug Administration-approved drugs that may be repurposed as NET inhibitors.
Collapse
Affiliation(s)
- Angela Meier
- Department of Anesthesiology, Division of Critical Care, University of California, San Diego School of Medicine, La Jolla
| | - George Sakoulas
- Division of Infectious Diseases, Sharp Rees-Stealy Medical Group, San Diego
- Collaborative to Halt Antibiotic-Resistant Microbes (CHARM)
| | - Victor Nizet
- Collaborative to Halt Antibiotic-Resistant Microbes (CHARM)
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego School of Medicine, La Jolla
| | - Erlinda R Ulloa
- Department of Pediatrics, University of California, Irvine School of Medicine
- Division of Infectious Disease, Children's Hospital of Orange County, Orange, California
| |
Collapse
|
8
|
Wang L, Huang FY, Dai SZ, Fu Y, Zhou X, Wang CC, Tan GH, Li Q. Progesterone modulates the immune microenvironment to suppress ovalbumin-induced airway inflammation by inhibiting NETosis. Sci Rep 2024; 14:17241. [PMID: 39060348 PMCID: PMC11282239 DOI: 10.1038/s41598-024-66439-6] [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: 12/15/2023] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Studies have demonstrated that prior to puberty, girls have a lower incidence and severity of asthma symptoms compared to boys. This study aimed to explore the role of progesterone (P4), a sex hormone, in reducing inflammation and altering the immune microenvironment in a mouse model of allergic asthma induced by OVA. Female BALB/c mice with or without ovariectomy to remove the influence of sex hormones were used for the investigations. Serum, bronchoalveolar lavage fluid (BALF), and lung tissue samples were collected for analysis. The results indicated that P4 treatment was effective in decreasing inflammation and mucus secretion in the lungs of OVA-induced allergic asthma mice. P4 treatment also reduced the influx of inflammatory cells into the BALF and increased the levels of Th1 and Th17 cytokines while decreasing the levels of Th2 and Treg cytokines in both BALF and lung microenvironment CD45+ T cells. Furthermore, P4 inhibited the infiltration of inflammatory cells into the lungs, suppressed NETosis, and reduced the number of pulmonary CD4+ T cells while increasing the number of regulatory T cells. The neutrophil elastase inhibitor GW311616A also suppressed airway inflammation and mucus production and modified the secretion of immune Th1, Th2, Th17, and Treg cytokines in lung CD45+ immune cells. These changes led to an alteration of the immunological milieu with increased Th1 and Th17 cells, accompanied by decreased Th2, Treg, and CD44+ T cells, similar to the effects of P4 treatment. Treatment with P4 inhibited NETosis by suppressing the p38 pathway activation, leading to reduced reactive oxygen species production. Moreover, P4 treatment hindered the release of double-stranded DNA during NETosis, thereby influencing the immune microenvironment in the lungs. These findings suggest that P4 treatment may be beneficial in reducing inflammation associated with allergic asthma by modulating the immune microenvironment. In conclusion, this research indicates the potential of P4 as a therapeutic agent for ameliorating inflammation in OVA-induced allergic asthma mice.
Collapse
Affiliation(s)
- Lin Wang
- Department of Respiratory Medicine, Hainan Province Clinical Medical Center of Respiratory Disease, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China
| | - Feng-Ying Huang
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, The Second Affiliated Hospital, Hainan Medical University, Haikou, 571199, Hainan, China.
| | - Shu-Zhen Dai
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, The Second Affiliated Hospital, Hainan Medical University, Haikou, 571199, Hainan, China
| | - Yongshu Fu
- Department of Respiratory Medicine, Hainan Province Clinical Medical Center of Respiratory Disease, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China
| | - Xiangdong Zhou
- Department of Respiratory Medicine, Hainan Province Clinical Medical Center of Respiratory Disease, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China
| | - Cai-Chun Wang
- Department of Respiratory Medicine, Hainan Province Clinical Medical Center of Respiratory Disease, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China.
| | - Guang-Hong Tan
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, The Second Affiliated Hospital, Hainan Medical University, Haikou, 571199, Hainan, China.
| | - Qi Li
- Department of Respiratory Medicine, Hainan Province Clinical Medical Center of Respiratory Disease, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China.
| |
Collapse
|
9
|
Aslanian-Kalkhoran L, Mehdizadeh A, Aghebati-Maleki L, Danaii S, Shahmohammadi-Farid S, Yousefi M. The role of neutrophils and neutrophil extracellular traps (NETs) in stages, outcomes and pregnancy complications. J Reprod Immunol 2024; 163:104237. [PMID: 38503075 DOI: 10.1016/j.jri.2024.104237] [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: 08/28/2023] [Revised: 01/23/2024] [Accepted: 03/07/2024] [Indexed: 03/21/2024]
Abstract
Neutrophils are the main components of innate immunity to eliminate infectious pathogens. Neutrophils play a role in several stages of the reproductive cycle, and their presence in the female reproductive system is highly regulated, so their function may change during pregnancy. Emerging evidence suggests that neutrophils are important at all stages of pregnancy, from implantation, placentation, and connective tissue regeneration to birth, as well as birth itself. Neutrophil extracellular traps (NETs) are defined as extracellular strands of unfolded DNA together with histone complexes and neutrophil granule proteins. NET formation is a new mechanism of these cells for their defense function. These strands containing DNA and antimicrobial peptides were initially recognized as one of the defense mechanisms of neutrophils, but later it was explained that they are involved in a variety of non-infectious diseases. Since the source of inflammation and tissue damage is the irregular activity of neutrophils, it is not surprising that NETosis are associated with a number of inflammatory conditions and diseases. The overexpression of NET components or non-principled NET clearance is associated with the risk of production and activation of autoantibodies, which results in participation in autoinflammatory and autoimmune disorders (SLE, RA), fibrosis, sepsis and other disorders such as vascular diseases, for example, thrombosis and atherosclerosis. Recent published articles have shown the role of neutrophils and extracellular traps (NETs) in pregnancy, childbirth and pregnancy-related diseases. The aim of this study was to identify and investigate the role of neutrophils and neutrophil extracellular traps (NETs) in the stages of pregnancy, as well as the complications caused by these cells.
Collapse
Affiliation(s)
- Lida Aslanian-Kalkhoran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Shahla Danaii
- Gynecology Department, Eastern Azerbaijan ACECR ART Centre, Eastern Azerbaijan Branch of ACECR, Tabriz, Iran
| | | | - Mehdi Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
10
|
Pastorek M, Konečná B, Janko J, Janovičová Ľ, Podracká Ľ, Záhumenský J, Šteňová E, Dúbrava M, Hodosy J, Vlková B, Celec P. Mitochondria-induced formation of neutrophil extracellular traps is enhanced in the elderly via Toll-like receptor 9. J Leukoc Biol 2023; 114:651-665. [PMID: 37648664 DOI: 10.1093/jleuko/qiad101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 09/01/2023] Open
Abstract
Neutrophil extracellular traps are potent antimicrobial weapons; however, their formation during sterile inflammation is detrimental, and the mechanism of induction is still unclear. Since advanced age is the primary clinical risk factor for poor outcomes in inflammatory diseases, we hypothesized that sterile stimuli, represented by mitochondria, would induce neutrophil extracellular trap formation in an age-dependent manner. Therefore, we analyzed induction of neutrophil extracellular traps in patients grouped according to age or immune status and observed that neutrophils from elderly patients responded to the presence of mitochondria with enhanced neutrophil extracellular trap formation. These neutrophil extracellular traps were also found to be more oxidized and exhibited higher resistance to DNase I degradation. Additionally, a higher concentration of residual neutrophil extracellular traps was detected in the plasma of the elderly. This plasma was capable of priming neutrophils through TLR9-mediated signaling, leading to further neutrophil extracellular trap formation, which was successfully inhibited with chloroquine. Finally, in a mouse model of mitochondria-induced acute lung injury, we observed that neutrophils from aged mice displayed impaired chemotactic activity but exhibited a trend of higher neutrophil extracellular trap formation. Thus, we propose that residual neutrophil extracellular traps circulating in the elderly preactivate neutrophils, making them more prone to enhanced neutrophil extracellular trap formation when exposed to mitochondria during sterile inflammation. Further investigation is needed to determine whether this vicious circle could be a suitable therapeutic target.
Collapse
Affiliation(s)
- Michal Pastorek
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Barbora Konečná
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Jakub Janko
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Ľubica Janovičová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Ľudmila Podracká
- Department of Pediatrics, Faculty of Medicine, Comenius University and National Institute of Children's Diseases, Limbová 1, 831 01 Bratislava, Slovakia
| | - Jozef Záhumenský
- 2nd Department of Gynecology and Obstetrics, Faculty of Medicine, University Hospital, Comenius University, Ružinovská 6, 821 06 Bratislava, Slovakia
| | - Emöke Šteňová
- 1st Department of Internal Medicine, Faculty of Medicine, University Hospital, Comenius University, Mickiewiczova 13, 813 69 Bratislava, Slovakia
| | - Martin Dúbrava
- 1st Department of Geriatrics, Faculty of Medicine, Comenius University, Limbová 5, 833 05 Bratislava, Slovakia
| | - Július Hodosy
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
- Department of Emergency Medicine Ružinov, Faculty of Medicine, University Hospital, Comenius University, Ružinovská 6, 821 06 Bratislava, Slovakia
| | - Barbora Vlková
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Peter Celec
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| |
Collapse
|
11
|
Mazur D, Satora M, Rekowska AK, Kabała Z, Łomża A, Kimber-Trojnar Ż, Leszczyńska-Gorzelak B. Influence of Breastfeeding on the State of Meta-Inflammation in Obesity-A Narrative Review. Curr Issues Mol Biol 2023; 45:9003-9018. [PMID: 37998742 PMCID: PMC10670570 DOI: 10.3390/cimb45110565] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
Obesity has become an emerging health issue worldwide that continues to grow in females of reproductive age as well. Obesity, as a multisystem and chronic disease, is associated with metabolic inflammation, which is defined as chronic low-grade systemic inflammation mediated by, i.a., adipose tissue macrophages. Lactation has been proven to have a beneficial influence on maternal health and could help restore metabolic balance, especially in the state of maternal obesity. In this review, we aimed to analyze the influence of breastfeeding on chronic low-grade meta-inflammation caused by obesity. We performed a comprehensive literature review using the PubMed, Science Direct, and Google Scholar electronic databases. For this purpose, we searched for "metabolic inflammation"; "meta-inflammation"; "obesity"; "breastfeeding"; "fetal programming"; "energy metabolism"; "postpartum"; "immunity"; "immune system"; and "inflammation" keyword combinations. While the clinical impact of breastfeeding on maternal and offspring health is currently well known, we decided to gain insight into more specific metabolic effects of adiposity, lipid, and glucose homeostasis, and immunological effects caused by the activity of cytokines, macrophages, and other immune system cells. Further research on the immunological and metabolic effects of breastfeeding in obese patients is key to understanding and potentially developing obesity therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | - Żaneta Kimber-Trojnar
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-059 Lublin, Poland; (D.M.); (M.S.); (A.K.R.); (Z.K.); (A.Ł.); (B.L.-G.)
| | | |
Collapse
|
12
|
Pham HTT, Magez S, Choi B, Baatar B, Jung J, Radwanska M. Neutrophil metalloproteinase driven spleen damage hampers infection control of trypanosomiasis. Nat Commun 2023; 14:5418. [PMID: 37669943 PMCID: PMC10480172 DOI: 10.1038/s41467-023-41089-w] [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: 01/13/2023] [Accepted: 08/18/2023] [Indexed: 09/07/2023] Open
Abstract
Recent blood transcriptomic analysis of rhodesiense sleeping sickness patients has revealed that neutrophil signature genes and activation markers constitute the top indicators of trypanosomiasis-associated inflammation. Here, we show that Trypanosoma brucei infection results in expansion and differentiation of four splenic neutrophil subpopulations, including Mki67+Birc5+Gfi1+Cebpe+ proliferation-competent precursors, two intermediate immature subpopulations and Cebpb+Spi1+Irf7+Mcl1+Csf3r+ inflammation reprogrammed mature neutrophils. Transcriptomic scRNA-seq profiling identified the largest immature subpopulation by Mmp8/9 positive tertiary granule markers. We confirmed the presence of both metalloproteinases in extracellular spleen homogenates and plasma. During infection, these enzymes digest extracellular matrix components in the absence of sufficient TIMP inhibitory activity, driving remodeling of the spleen follicular architecture. Neutrophil depletion prevents the occurrence of organ damage, resulting in increased plasma cell numbers and prolonged host survival. We conclude that trypanosomiasis-associated neutrophil activation is a major contributor to the destruction of the secondary lymphoid architecture, required for maintaining an efficient adaptive immune response.
Collapse
Affiliation(s)
- Hien Thi Thu Pham
- Laboratory for Biomedical Research, Department of Environmental Technology, Food Technology and Molecular Biotechnology KR01, Ghent University Global Campus, Incheon, South Korea
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Stefan Magez
- Laboratory for Biomedical Research, Department of Environmental Technology, Food Technology and Molecular Biotechnology KR01, Ghent University Global Campus, Incheon, South Korea
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Boyoon Choi
- Laboratory for Biomedical Research, Department of Environmental Technology, Food Technology and Molecular Biotechnology KR01, Ghent University Global Campus, Incheon, South Korea
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Bolortsetseg Baatar
- Laboratory for Biomedical Research, Department of Environmental Technology, Food Technology and Molecular Biotechnology KR01, Ghent University Global Campus, Incheon, South Korea
| | - Joohee Jung
- Duksung Women's University, Seoul, South Korea
| | - Magdalena Radwanska
- Laboratory for Biomedical Research, Department of Environmental Technology, Food Technology and Molecular Biotechnology KR01, Ghent University Global Campus, Incheon, South Korea.
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
13
|
Li W, Wang Z, Su C, Liao Z, Pei Y, Wang J, Li Z, Fu S, Liu J. The effect of neutrophil extracellular traps in venous thrombosis. Thromb J 2023; 21:67. [PMID: 37328882 DOI: 10.1186/s12959-023-00512-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 06/11/2023] [Indexed: 06/18/2023] Open
Abstract
Neutrophil extracellular traps (NETs) as special release products of neutrophils have received extensive attention. They are composed of decondensed chromatin and coated with nucleoproteins, including histones and some granulosa proteins. NETs can form a network structure to effectively capture and eliminate pathogens and prevent their spread. Not only that, recent studies have shown that NETs also play an important role in venous thrombosis. This review provides the most important updated evidence regarding the mechanism of NETs formation and the role of NETs in the process of venous thrombosis. The potential prophylactic and therapeutic value of NETs in venous thrombotic disease will also be discussed.
Collapse
Affiliation(s)
- Weiwei Li
- Department of Hepatobiliary Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, 067000, China
| | - Zixiang Wang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, 067000, China
| | - Chen'guang Su
- Department of Hepatobiliary Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, 067000, China
| | - Zheng Liao
- Department of Hepatobiliary Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, 067000, China
| | - Yinxuan Pei
- Department of Hepatobiliary Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, 067000, China
| | - Jianli Wang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, 067000, China
| | - Zixin Li
- Department of Pathology, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, 067000, China
| | - Shijie Fu
- Department of Orthopedic, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, 067000, China
| | - Jinlong Liu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei Province, 067000, China.
| |
Collapse
|
14
|
Motomura K, Miller D, Galaz J, Liu TN, Romero R, Gomez-Lopez N. The effects of progesterone on immune cellular function at the maternal-fetal interface and in maternal circulation. J Steroid Biochem Mol Biol 2023; 229:106254. [PMID: 36681283 PMCID: PMC10038932 DOI: 10.1016/j.jsbmb.2023.106254] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 01/16/2023] [Accepted: 01/16/2023] [Indexed: 01/19/2023]
Abstract
Progesterone is a sex steroid hormone that plays a critical role in the establishment and maintenance of pregnancy. This hormone drives numerous maternal physiological adaptations to ensure the continuation of pregnancy and to facilitate fetal growth, including broad and potent modulation of the maternal immune system to promote maternal-fetal tolerance. In this brief review, we provide an overview of the immunomodulatory functions of progesterone in the decidua, placenta, myometrium, and maternal circulation during pregnancy. Specifically, we summarize current evidence of the regulated functions of innate and adaptive immune cells induced by progesterone and its downstream effector molecules in these compartments, including observations in human pregnancy and in animal models. Our review highlights the gaps in knowledge of interactions between progesterone and maternal cellular immunity that may direct future research.
Collapse
Affiliation(s)
- Kenichiro Motomura
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Derek Miller
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jose Galaz
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Tzu Ning Liu
- Wayne State University School of Medicine, Detroit, MI, USA
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA; Detroit Medical Center, Detroit, MI, USA
| | - Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD, and Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA; Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
15
|
Tariq MB, Lee J, McCullough LD. Sex differences in the inflammatory response to stroke. Semin Immunopathol 2023; 45:295-313. [PMID: 36355204 PMCID: PMC10924671 DOI: 10.1007/s00281-022-00969-x] [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: 07/29/2022] [Accepted: 10/18/2022] [Indexed: 11/11/2022]
Abstract
Ischemic stroke is a leading cause of morbidity and mortality and disproportionally affects women, in part due to their higher longevity. Older women have poorer outcomes after stroke with high rates of cognitive deficits, depression, and reduced quality of life. Post-stroke inflammatory responses are also sexually dimorphic and drive differences in infarct size and recovery. Factors that influence sex-specific immune responses can be both intrinsic and extrinsic. Differences in gonadal hormone exposure, sex chromosome compliment, and environmental/social factors can drive changes in transcriptional and metabolic profiles. In addition, how these variables interact, changes across the lifespan. After the onset of ischemic injury, necrosis and apoptosis occur, which activate microglia and other glial cells within the central nervous system, promoting the release of cytokines and chemokines and neuroinflammation. Cells involved in innate and adaptive immune responses also have dual functions after stroke as they can enhance inflammation acutely, but also contribute to suppression of the inflammatory cascade and later repair. In this review, we provide an overview of the current literature on sex-specific inflammatory responses to ischemic stroke. Understanding these differences is critical to identifying therapeutic options for both men and women.
Collapse
Affiliation(s)
- Muhammad Bilal Tariq
- Memorial Hermann Hospital-Texas Medical Center, Houston, TX, 77030, USA
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin St, MSB7044B, Houston, TX, 77030, USA
| | - Juneyoung Lee
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin St, MSB7044B, Houston, TX, 77030, USA
| | - Louise D McCullough
- Memorial Hermann Hospital-Texas Medical Center, Houston, TX, 77030, USA.
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin St, MSB7044B, Houston, TX, 77030, USA.
| |
Collapse
|
16
|
Chakraborty B, Byemerwa J, Krebs T, Lim F, Chang CY, McDonnell DP. Estrogen Receptor Signaling in the Immune System. Endocr Rev 2023; 44:117-141. [PMID: 35709009 DOI: 10.1210/endrev/bnac017] [Citation(s) in RCA: 101] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Indexed: 01/14/2023]
Abstract
The immune system functions in a sexually dimorphic manner, with females exhibiting more robust immune responses than males. However, how female sex hormones affect immune function in normal homeostasis and in autoimmunity is poorly understood. In this review, we discuss how estrogens affect innate and adaptive immune cell activity and how dysregulation of estrogen signaling underlies the pathobiology of some autoimmune diseases and cancers. The potential roles of the major circulating estrogens, and each of the 3 estrogen receptors (ERα, ERβ, and G-protein coupled receptor) in the regulation of the activity of different immune cells are considered. This provides the framework for a discussion of the impact of ER modulators (aromatase inhibitors, selective estrogen receptor modulators, and selective estrogen receptor downregulators) on immunity. Synthesis of this information is timely given the considerable interest of late in defining the mechanistic basis of sex-biased responses/outcomes in patients with different cancers treated with immune checkpoint blockade. It will also be instructive with respect to the further development of ER modulators that modulate immunity in a therapeutically useful manner.
Collapse
Affiliation(s)
- Binita Chakraborty
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jovita Byemerwa
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Taylor Krebs
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA.,Known Medicine, Salt Lake City, UT 84108, USA
| | - Felicia Lim
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ching-Yi Chang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
17
|
Guillotin F, Fortier M, Portes M, Demattei C, Mousty E, Nouvellon E, Mercier E, Chea M, Letouzey V, Gris JC, Bouvier S. Vital NETosis vs. suicidal NETosis during normal pregnancy and preeclampsia. Front Cell Dev Biol 2023; 10:1099038. [PMID: 36684420 PMCID: PMC9849884 DOI: 10.3389/fcell.2022.1099038] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/20/2022] [Indexed: 01/07/2023] Open
Abstract
Background: NETosis occurs in the context of infection or inflammation and results in the expulsion of decondensed DNA filaments called NETs (Neutrophil Extracellular Traps) into the extracellular environment. NETosis activates coagulation and contributes to the thrombotic risk of inflammatory diseases. To date, two mechanisms of NETosis have been identified: suicidal NETosis, in which neutrophils die after expelling the filaments; and vital NETosis, in which expulsion appears without altering the membrane. Human pregnancy is associated with a mild pro-inflammatory state, which is increased in the event of complications such as preeclampsia (PE). NETosis has been observed in these situations, but the mechanism of its production has not yet been studied. The aim of our study was to evaluate the balance of vital vs. suicidal NETosis in normal pregnancy and in PE. Patients/Methods: Neutrophils from healthy volunteers were stimulated with plasma from normal pregnancies (n = 13) and from women developing preeclampsia (n = 13). Immunofluorescent labelling was performed to determine the percentages and origin of NETs in both groups. Inhibition with suicidal or vital NETosis inhibitors was also performed to validate our results. Results: We found a significant increase in NETs in women with PE compared to women with normal pregnancies. We showed that vital and non-vital NETosis are present in normal and preeclamptic pregnancies. We demonstrated that the higher proportion of NETs observed in PE was due to non-vital NETosis whose main component is represented by suicidal NETosis. Discussion: These results suggest the important part of non-vital NETosis in the pathophysiology of PE.
Collapse
Affiliation(s)
| | - Mathieu Fortier
- Department of Haematology, University Hospital, Nîmes, France
| | - Marie Portes
- Department of Gynecology and Obstetrics, University Hospital, Nîmes, France
| | - Christophe Demattei
- Department of Biostatistics, Public Health and Innovation in Methodology, Nîmes University Hospital, Nîmes, France
| | - Eve Mousty
- Department of Gynecology and Obstetrics, University Hospital, Nîmes, France
| | - Eva Nouvellon
- Department of Haematology, University Hospital, Nîmes, France,UA11 INSERM—UM Institut Desbrest d’Épidémiologie et de Santé Publique (IDESP), Montpellier, France
| | - Eric Mercier
- Department of Haematology, University Hospital, Nîmes, France,UA11 INSERM—UM Institut Desbrest d’Épidémiologie et de Santé Publique (IDESP), Montpellier, France,Faculty of Pharmaceutical and Biological Sciences, Montpellier University, France
| | - Mathias Chea
- Department of Haematology, University Hospital, Nîmes, France
| | - Vincent Letouzey
- Department of Gynecology and Obstetrics, University Hospital, Nîmes, France,Department of artificial polymers, Max Mousseron Institute of Biomolecules, CNRS UMR 5247, Univ Montpellier, Montpellier, France
| | - Jean-Christophe Gris
- Department of Haematology, University Hospital, Nîmes, France,UA11 INSERM—UM Institut Desbrest d’Épidémiologie et de Santé Publique (IDESP), Montpellier, France,Faculty of Pharmaceutical and Biological Sciences, Montpellier University, France,I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Sylvie Bouvier
- Department of Haematology, University Hospital, Nîmes, France,UA11 INSERM—UM Institut Desbrest d’Épidémiologie et de Santé Publique (IDESP), Montpellier, France,Faculty of Pharmaceutical and Biological Sciences, Montpellier University, France,*Correspondence: Sylvie Bouvier,
| |
Collapse
|
18
|
Ye H, Li L, Dong Y, Zheng Q, Sha Y, Li L, Yang P, Jia Y, Gu J. Dysregulated low-density granulocyte contributes to early spontaneous abortion. Front Immunol 2023; 14:1119756. [PMID: 36911722 PMCID: PMC9995479 DOI: 10.3389/fimmu.2023.1119756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Spontaneous abortion (SA) is a common adverse pregnancy event with unclarified pathogenesis and limited therapeutic efficiency. Although most SA cases with the euploid embryo(s) are associated with immunological factors, the contribution of low-density granulocyte (LDG) in SA pathogenesis is rarely reported. This study aimed to investigate the serial characteristics and possible contribution of LDG and their subpopulations in early pregnancy, especially in early SA. Unpregnant (UP), normally pregnant (NP), and SA women were recruited, and the peripheral blood and endometrium/decidua were collected for LDG isolation and histological observation. The percentage, phenotype, and subpopulations of LDG were analyzed via flow cytometric analysis, and the ability of Nets formation was assessed by immunofluorescent and immunohistochemical assays. As a result, 43 participants were enrolled, including 10 UP, 15 NP, and 18 SA women. Compared with the UP group, the LDG percentage in peripheral blood mononuclear cells (PBMCs) and decidual immune cells (DICs) increased in the NP group, while the loss of this increase was observed in the SA group. Meanwhile, CD16int/- cell percentage in peripheral blood LDG (PB-LDG) increased in the NP and SA groups, and insufficient activation of CD16hi PB-LDG characterized by reduced CD11b expression was discovered in the SA group. Moreover, the LDG percentage in DICs was higher than that in PBMCs, and the decidual LDG (D-LDG) showed a surface marker expression profile that is easier to be activated in the pregnant cohort (NP + SA women). Finally, increased decidual Nets formation was observed in the SA group compared with the NP group, and more Nets formation was detected in D-LDG of NP and SA women following PMA stimulation. Overall, LDG participates in the maintenance of early pregnancy, while dysregulated LDG is responsible for early SA, providing novel potential targets for further exploration of SA pathogenesis and therapeutics.
Collapse
Affiliation(s)
- Hongxia Ye
- Department of Reproductive Immunology, Chengdu Xi’nan Gynecology Hospital, Chengdu, Sichuan, China
- Department of Reproductive Immunology, Chengdu Jinjiang Hospital for Maternal & Child Health Care, Chengdu, Sichuan, China
- Jinxin Research Institute for Reproductive Medicine and Genetics, Chengdu Xi’nan Gynecology Hospital, Chengdu, Sichuan, China
| | - Lan Li
- Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yajun Dong
- Department of Reproductive Immunology, Chengdu Xi’nan Gynecology Hospital, Chengdu, Sichuan, China
| | - Qu Zheng
- Department of Laboratory Medicine, Chengdu Xi’nan Gynecology Hospital, Chengdu, Sichuan, China
| | - Yulin Sha
- Jinxin Research Institute for Reproductive Medicine and Genetics, Chengdu Xi’nan Gynecology Hospital, Chengdu, Sichuan, China
| | - Li Li
- Department of Gynecology, Sichuan Jinxin Women & Children Hospital, Chengdu, Sichuan, China
| | - Panyu Yang
- Department of Laboratory Medicine, Chengdu Xi’nan Gynecology Hospital, Chengdu, Sichuan, China
| | - Yan Jia
- Department of Reproductive Immunology, Chengdu Xi’nan Gynecology Hospital, Chengdu, Sichuan, China
- *Correspondence: Yan Jia, ; Jiang Gu,
| | - Jiang Gu
- Jinxin Research Institute for Reproductive Medicine and Genetics, Chengdu Xi’nan Gynecology Hospital, Chengdu, Sichuan, China
- Department of Pathology and Pathophysiology, Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Shantou University Medical College, Shantou, China
- *Correspondence: Yan Jia, ; Jiang Gu,
| |
Collapse
|
19
|
Christensen AO, Li G, Young CH, Snow B, Khan SA, DeVore SB, Edwards S, Bouma GJ, Navratil AM, Cherrington BD, Rothfuss HM. Peptidylarginine deiminase enzymes and citrullinated proteins in female reproductive physiology and associated diseases†. Biol Reprod 2022; 107:1395-1410. [PMID: 36087287 PMCID: PMC10248218 DOI: 10.1093/biolre/ioac173] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/30/2022] [Accepted: 09/07/2022] [Indexed: 09/15/2023] Open
Abstract
Citrullination, the post-translational modification of arginine residues, is catalyzed by the four catalytically active peptidylarginine deiminase (PAD or PADI) isozymes and alters charge to affect target protein structure and function. PADs were initially characterized in rodent uteri and, since then, have been described in other female tissues including ovaries, breast, and the lactotrope and gonadotrope cells of the anterior pituitary gland. In these tissues and cells, estrogen robustly stimulates PAD expression resulting in changes in levels over the course of the female reproductive cycle. The best-characterized targets for PADs are arginine residues in histone tails, which, when citrullinated, alter chromatin structure and gene expression. Methodological advances have allowed for the identification of tissue-specific citrullinomes, which reveal that PADs citrullinate a wide range of enzymes and structural proteins to alter cell function. In contrast to their important physiological roles, PADs and citrullinated proteins are also involved in several female-specific diseases including autoimmune disorders and reproductive cancers. Herein, we review current knowledge regarding PAD expression and function and highlight the role of protein citrullination in both normal female reproductive tissues and associated diseases.
Collapse
Affiliation(s)
- Amanda O Christensen
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY 82071, USA
| | - Guangyuan Li
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY 82071, USA
| | - Coleman H Young
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY 82071, USA
| | - Bryce Snow
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY 82071, USA
| | | | - Stanley B DeVore
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Sydney Edwards
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Gerrit J Bouma
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Amy M Navratil
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY 82071, USA
| | - Brian D Cherrington
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY 82071, USA
| | - Heather M Rothfuss
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY 82071, USA
| |
Collapse
|
20
|
Collins MK, McCutcheon CR, Petroff MG. Impact of Estrogen and Progesterone on Immune Cells and Host–Pathogen Interactions in the Lower Female Reproductive Tract. THE JOURNAL OF IMMUNOLOGY 2022; 209:1437-1449. [DOI: 10.4049/jimmunol.2200454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/05/2022] [Indexed: 11/05/2022]
|
21
|
Mohammed S, Alhussien MN, Dang AK. Pregnancy stage-dependent modulation of neutrophil function may impact embryo survivability and pregnancy outcome in crossbred cows. Theriogenology 2022; 191:200-206. [PMID: 35998403 DOI: 10.1016/j.theriogenology.2022.08.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/26/2022]
Abstract
Pregnancy is a complicated physiological process that involves synchronized coordination between immune and endocrine systems. Neutrophils have been suggested as a critical immune cell for embryo implantation and pregnancy maintenance. The present study was conducted to evaluate the dynamic changes in the mRNA expressions of the cluster of designation (CD11b, CD31, CD44 and CD62L) molecules and interferon-stimulated genes (ISG15, MX1 and OAS1) in blood neutrophils throughout pregnancy in dairy cows and correlate them with the outcome of pregnancy. Blood samples were taken from negative control (NC) group, and non-pregnant (NP) group at the time of artificial insemination (AI, day zero) and on days 10, 14, 16, 18, and 21 post-AI. In pregnant (P) cows, samples were taken as described above and after every 30 days until the time of parturition. In aborted cows, samples were collected until the time of the abortion. Comparison between pregnant, non-pregnant and aborted cows revealed that the expression of CD molecules increased (p < 0.05) on days 14, 16, 18 and 21 post-AI only in NP cows as compared to other groups. Although the expression of CD molecules remained constant throughout the study period in pregnant and aborted cows, the expression of CD11b, CD31 and CD62L increased (p < 0.05) on the day of abortion and parturition. Unlike CD molecules, the expression of CD44 decreased significantly (p < 0.05) at the time of abortion. There was a significant (p < 0.05) increase in the expression of interferon-stimulated genes including MX1, OAS1 and ISG15 during the peri-implantation period in pregnant cows, and at the time of abortion in aborted cows. However, the expression of ISGs was lower (p < 0.05) in non-pregnant cows as compared to the other groups. The results revealed the critical role played by neutrophils during pregnancy and form the basis to unravel the underlying mechanism for neutrophil associated immunological infertility in bovines.
Collapse
Affiliation(s)
- Seid Mohammed
- Bio and Emerging Technology Institute, Addis Ababa, Ethiopia; Lactation and Immuno-Physiology Laboratory, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India
| | - Mohanned Naif Alhussien
- Lactation and Immuno-Physiology Laboratory, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India; Reproductive Biotechnology, TUM School of Life Sciences Weihenstephan, Technical University Munich, 85354, Freising, Germany.
| | - Ajay Kumar Dang
- Lactation and Immuno-Physiology Laboratory, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India
| |
Collapse
|
22
|
Pastorek M, Drobná D, Celec P. Could neutrophil extracellular traps drive the development of autism? Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
23
|
Hebeda CB, Savioli AC, Scharf P, de Paula-Silva M, Gil CD, Farsky SHP, Sandri S. Neutrophil depletion in the pre-implantation phase impairs pregnancy index, placenta and fetus development. Front Immunol 2022; 13:969336. [PMID: 36248911 PMCID: PMC9558710 DOI: 10.3389/fimmu.2022.969336] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Maternal neutrophils cells are players in gestational tolerance and fetus delivery. Nonetheless, their actions in each phase of the pregnancy are unknown. We here investigated the role of maternal neutrophil depletion before the blastocyst implantation phase and outcomes in the pregnancy index, placenta, and fetus development. Neutrophils were pharmacologically depleted by i.p. injection of anti-Gr1 (anti-neutrophils; 200 µg) 24 hours after plug visualization in allogeneic-mated C57BL/6/BALB/c mice. Depletion of peripheral neutrophils lasted until 48 hours after anti-Gr1 injection (gestational day 1.5-3.5). On gestational day 5.5, neutrophil depletion impaired the blastocyst implantation, as 50% of pregnant mice presented reduced implantation sites. On gestational day 18.5, neutrophil depletion reduced the pregnancy rate and index, altered the placenta disposition in the uterine horns, and modified the structure of the placenta, detected by reduced junctional zone, associated with decreased numbers of giant trophoblast cells, spongiotrophoblast. Reduced number of placenta cells labeled for vascular endothelial growth factor (VEGF), platelet-endothelial cell adhesion molecule (PECAM-1), and intercellular cell adhesion molecule (ICAM-1), important markers of angiogenesis and adhesiveness, were detected in neutrophil depleted mice. Furthermore, neutrophil depletion promoted a higher frequency of monocytes, natural killers, and T regulatory cells, and lower frequency of cytotoxic T cells in the blood, and abnormal development of offspring. Associated data obtained herein highlight the pivotal role of neutrophils actions in the early stages of pregnancy, and address further investigations on the imbricating signaling evoked by neutrophils in the trophoblastic interaction with uterine epithelium.
Collapse
Affiliation(s)
- Cristina Bichels Hebeda
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, SP, São Paulo, Brazil
- Núcleo de Pesquisa em Ciências Médicas, Fundação Universidade para o Desenvolvimento do Alto Vale do Itajaí – UNIDAVI, Rio do Sul, SC, Brazil
| | - Anna Carolina Savioli
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, SP, São Paulo, Brazil
| | - Pablo Scharf
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, SP, São Paulo, Brazil
| | - Marina de Paula-Silva
- Center for Stem Cells and Regenerative Medicine, King’s College London, London, United Kingdom
| | - Cristiane Damas Gil
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Sandra Helena Poliselli Farsky
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, SP, São Paulo, Brazil
| | - Silvana Sandri
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, SP, São Paulo, Brazil
- *Correspondence: Silvana Sandri,
| |
Collapse
|
24
|
Elevated G-CSF, IL8, and HGF in patients with definite Meniere's disease may indicate the role of NET formation in triggering autoimmunity and autoinflammation. Sci Rep 2022; 12:16309. [PMID: 36175465 PMCID: PMC9522806 DOI: 10.1038/s41598-022-20774-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/19/2022] [Indexed: 11/08/2022] Open
Abstract
The etiology and mechanism causing Meniere’s disease (MD) are not understood. The present study investigated the possible molecular mechanism of autoimmunity and autoinflammation associated with MD. Thirty-eight patients with definite MD and 39 normal volunteers were recruited, and 48 human cytokines/chemokines were quantified. In patients with MD pure tone audiograms, tympanograms and standard blood tests were performed. The mean hearing loss in the worse ear was 44.1 dB nHL. Compared to the referents, the concentrations of TNFα, IL1α, IL8, CTACK, MIP1α, MIP1β, G-CSF, and HGF in the sera of patients with MD were significantly elevated, while those of TRAIL and PDGFBB were significantly decreased. The area under the receiver operating characteristic curve (AUC) showed that G-CSF, MIP1α, and IL8 were above 0.8 and could be used to diagnose MD (p < 0.01), and the AUCs of CTACK and HGF were above 0.7 and acceptable to discriminate the MD group from the control group (p < 0.01). The revised AUCs (1 − AUC) of TRAIL and PDGFBB were above 0.7 and could also be used in the diagnosis of MD (p < 0.01). The linear regression showed significant correlations between MIP1α and GCSF, between IL2Rα and GCSF, between IL8 and HGF, between MIP1α and IL8, and between SCF and CTACK; there was a marginal linear association between IP10 and MIP1α. Linear regression also showed that there were significant age-related correlations of CTACK and MIG expression in the MD group (p < 0.01, ANOVA) but not in the control group. We hypothesize that G-CSF, IL8, and HGF, which are involved in the development of neutrophil extracellular traps (NETs) and through various mechanisms influence the functions of macrophages, lymphocytes, and dendritic cells, among others, are key players in the development of EH and MD and could be useful in elucidating the pathophysiological mechanisms leading to MD. Biomarkers identified in the present study may suggest that both autoimmune and autoinflammatory mechanisms are involved in MD. In the future, it will be valuable to develop a cost-effective method to detect G-CSF, IL8, HGF, CTACK, MIP1α, TRAIL, and PDGFBB in the serum of patient that have diagnostic relevance.
Collapse
|
25
|
Hu W, Lee SML, Bazhin AV, Guba M, Werner J, Nieß H. Neutrophil extracellular traps facilitate cancer metastasis: cellular mechanisms and therapeutic strategies. J Cancer Res Clin Oncol 2022; 149:2191-2210. [PMID: 36050539 PMCID: PMC9436160 DOI: 10.1007/s00432-022-04310-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/15/2022] [Indexed: 11/24/2022]
Abstract
Background The formation of neutrophil extracellular traps (NETs) was initially discovered as a novel immune response against pathogens. Recent studies have also suggested that NETs play an important role in tumor progression. This review summarizes the cellular mechanisms by which NETs promote distant metastasis and discusses the possible clinical applications targeting NETs. Method The relevant literature from PubMed and Google Scholar (2001–2021) have been reviewed for this article. Results The presence of NETs has been detected in various primary tumors and metastatic sites. NET-associated interactions have been observed throughout the different stages of metastasis, including initial tumor cell detachment, intravasation and extravasation, the survival of circulating tumor cells, the settlement and the growth of metastatic tumor cells. Several in vitro and in vivo studies proved that inhibiting NET formation resulted in anti-cancer effects. The biosafety and efficacy of some NET inhibitors have also been demonstrated in early phase clinical trials. Conclusions Considering the role of NETs in tumor progression, NETs could be a promising diagnostic and therapeutic target for cancer management. However, current evidence is mostly derived from experimental models and as such more clinical studies are still needed to verify the clinical significance of NETs in oncological settings.
Collapse
Affiliation(s)
- Wenxing Hu
- Department of General, Visceral, and Transplant Surgery, University Hospital, Ludwig-Maximilians-University, Marchioninistr. 15, 81377, Munich, Germany
| | - Serene M L Lee
- Department of General, Visceral, and Transplant Surgery, University Hospital, Ludwig-Maximilians-University, Marchioninistr. 15, 81377, Munich, Germany
| | - Alexandr V Bazhin
- Department of General, Visceral, and Transplant Surgery, University Hospital, Ludwig-Maximilians-University, Marchioninistr. 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Markus Guba
- Department of General, Visceral, and Transplant Surgery, University Hospital, Ludwig-Maximilians-University, Marchioninistr. 15, 81377, Munich, Germany
| | - Jens Werner
- Department of General, Visceral, and Transplant Surgery, University Hospital, Ludwig-Maximilians-University, Marchioninistr. 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.,Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Hanno Nieß
- Department of General, Visceral, and Transplant Surgery, University Hospital, Ludwig-Maximilians-University, Marchioninistr. 15, 81377, Munich, Germany.
| |
Collapse
|
26
|
Sex Steroids Effects on Asthma: A Network Perspective of Immune and Airway Cells. Cells 2022; 11:cells11142238. [PMID: 35883681 PMCID: PMC9318292 DOI: 10.3390/cells11142238] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/13/2022] [Accepted: 07/17/2022] [Indexed: 11/17/2022] Open
Abstract
A multitude of evidence has suggested the differential incidence, prevalence and severity of asthma between males and females. A compilation of recent literature recognized sex differences as a significant non-modifiable risk factor in asthma pathogenesis. Understanding the cellular and mechanistic basis of sex differences remains complex and the pivotal point of this ever elusive quest, which remains to be clarified in the current scenario. Sex steroids are an integral part of human development and evolution while also playing a critical role in the conditioning of the immune system and thereby influencing the function of peripheral organs. Classical perspectives suggest a pre-defined effect of sex steroids, generalizing estrogens popularly under the “estrogen paradox” due to conflicting reports associating estrogen with a pro- and anti-inflammatory role. On the other hand, androgens are classified as “anti-inflammatory,” serving a protective role in mitigating inflammation. Although considered mainstream and simplistic, this observation remains valid for numerous reasons, as elaborated in the current review. Women appear immune-favored with stronger and more responsive immune elements than men. However, the remarkable female predominance of diverse autoimmune and allergic diseases contradicts this observation suggesting that hormonal differences between the sexes might modulate the normal and dysfunctional regulation of the immune system. This review illustrates the potential relationship between key elements of the immune cell system and their interplay with sex steroids, relevant to structural cells in the pathophysiology of asthma and many other lung diseases. Here, we discuss established and emerging paradigms in the clarification of observed sex differences in asthma in the context of the immune system, which will deepen our understanding of asthma etiopathology.
Collapse
|
27
|
Bitsadze V, Khizroeva J, Alexander M, Elalamy I. Venous thrombosis risk factors in pregnant women. J Perinat Med 2022; 50:505-518. [PMID: 35044114 DOI: 10.1515/jpm-2022-0008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/07/2022] [Indexed: 11/15/2022]
Abstract
Venous thromboembolism (VTE) is the third most common cause of death on Earth after myocardial infarctions and strokes, according to the World Health Organization (WHO). Pregnancy is a unique condition of woman, when enormous changes occur in functioning of the most important systems of homeostasis in a relatively short time. These are physiological hypercoagulation, slowing of blood flow, increase in circulating blood volume, etc. However, while being physiological, these changes increase the risks of venous thromboembolism by almost 6 times. In some cases, there appears an imbalance or dissociation between the functioning of natural antithrombotic systems and the activation of coagulation as a consequence of genetically or acquired determined causes (genetic thrombophilia, antiphospholipid syndrome, comorbidities, obstetric complications and other exogenous and endogenous factors). Accordingly, identification of risk factors, their systematization, and determination of VTE risks in pregnancy and puerperium is one of the most important tasks of clinical medicine. Various recommendations have appeared for practitioners during the last 10-15 years on the basis of the risk factors analysis in order to prevent VTE in pregnant women more effectively. Nevertheless, none of these recommendations can yet take into account all risk factors, although convenient scoring systems have emerged for risk assessment and clear recommendations on anti-thrombotic prophylaxis regimens in risk groups in recent years. This article will review historical understanding of thrombosis in pregnant women, progress in understanding VTE risk factors in pregnant women, and available reserves in identifying new risk factors during pregnancy and puerperium in order to stratify risks more efficiently.
Collapse
Affiliation(s)
- Victoria Bitsadze
- Department of Obstetrics and Gynecology, Russian Academy of Sciences, I.M.Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Jamilya Khizroeva
- Department of Obstetrics and Gynecology, Russian Academy of Sciences, I.M.Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Makatsariya Alexander
- Department of Obstetrics and Gynecology, Russian Academy of Sciences, I.M.Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Ismail Elalamy
- Department Hematology and Thrombosis Center, Medicine, Sorbonne University, Paris, France
| |
Collapse
|
28
|
Rosenzweig R, Kumar V, Gupta S, Bermeo-Blanco O, Stratton MS, Gumina RJ, Bansal SS. Estrogen Receptor-β Agonists Modulate T-Lymphocyte Activation and Ameliorate Left Ventricular Remodeling During Chronic Heart Failure. Circ Heart Fail 2022; 15:e008997. [PMID: 35730443 DOI: 10.1161/circheartfailure.121.008997] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND CD4+ T cells temporally transition from protective to pathological during ischemic heart failure (HF; 8 weeks postmyocardial infarction). Cellular mechanisms mediating this shift are unknown. METHODS RNA-sequencing of cardiac CD4+ T cells and flow cytometric analysis of immune cells was conducted. RESULTS RNA-sequencing of CD4+ T cells from the failing hearts of male mice indicated activation of ER (estrogen receptor)-α signaling. Flow cytometric analysis showed that ERα in CD4+ T cells decreases significantly at 3-day postmyocardial infarction but increases during HF. To antagonize ERα, we tested a novel ERβ agonist (OSU-ERb-012) to inhibit T cells and blunt left ventricular remodeling. Proliferation assays showed that OSU-ERb-012 dose-dependently inhibited proliferation and proinflammatory cytokine expression in anti-CD3/CD28 stimulated splenic T cells isolated from both the sexes. For in vivo efficacy, 10- to 12-week-old male and ovariectomized female mice were randomized at 4 weeks postmyocardial infarction and treated with either vehicle or drug (60 mg/kg per day; oral). While vehicle-treated HF mice displayed progressive left ventricular dilatation with significantly increased end-systolic and end-diastolic volumes from 4 to 8 weeks postmyocardial infarction, treatment with OSU-ERb-012 significantly blunted these changes and stopped left ventricular remodeling in both the sexes. Reduction in tibia-normalized heart and left ventricular weights, cardiomyocyte hypertrophy and interstitial fibrosis further supported these results. Additionally, OSU-ERb-012 treatment selectively inhibited cardiac, splenic, and circulating CD4+ T cells without affecting other myeloid and lymphoid cells in the HF mice. CONCLUSIONS Our studies indicate that ERβ agonists and OSU-ERb-012, in particular, could be used as selective immunomodulatory drugs to inhibit CD4+ T cells during chronic HF.
Collapse
Affiliation(s)
- Rachel Rosenzweig
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus. (R.R., V.K., S.G., O.B.-B., M.S.S., R.J.G., S.S.B.).,The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus. (R.R., V.K., S.G., O.B.-B., M.S.S., R.J.G., S.S.B.)
| | - Vinay Kumar
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus. (R.R., V.K., S.G., O.B.-B., M.S.S., R.J.G., S.S.B.).,The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus. (R.R., V.K., S.G., O.B.-B., M.S.S., R.J.G., S.S.B.)
| | - Sahil Gupta
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus. (R.R., V.K., S.G., O.B.-B., M.S.S., R.J.G., S.S.B.).,The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus. (R.R., V.K., S.G., O.B.-B., M.S.S., R.J.G., S.S.B.)
| | - Oscar Bermeo-Blanco
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus. (R.R., V.K., S.G., O.B.-B., M.S.S., R.J.G., S.S.B.).,The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus. (R.R., V.K., S.G., O.B.-B., M.S.S., R.J.G., S.S.B.)
| | - Matthew S Stratton
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus. (R.R., V.K., S.G., O.B.-B., M.S.S., R.J.G., S.S.B.).,The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus. (R.R., V.K., S.G., O.B.-B., M.S.S., R.J.G., S.S.B.)
| | - Richard J Gumina
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus. (R.R., V.K., S.G., O.B.-B., M.S.S., R.J.G., S.S.B.).,The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus. (R.R., V.K., S.G., O.B.-B., M.S.S., R.J.G., S.S.B.).,Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus. (R.J.G., S.S.B.)
| | - Shyam S Bansal
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus. (R.R., V.K., S.G., O.B.-B., M.S.S., R.J.G., S.S.B.).,The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus. (R.R., V.K., S.G., O.B.-B., M.S.S., R.J.G., S.S.B.).,Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus. (R.J.G., S.S.B.)
| |
Collapse
|
29
|
Hauk V, D'Eramo L, Calo G, Merech F, Doga L, Lara B, Gliosca L, Massone C, Molgatini S, Ramhorst R, Squassi A, Pérez Leirós C. Gingival crevicular fluid from pregnant women impairs trophoblast cell function and trophoblast-neutrophil interaction. Am J Reprod Immunol 2022; 88:e13558. [PMID: 35511077 DOI: 10.1111/aji.13558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/10/2022] [Accepted: 04/19/2022] [Indexed: 12/15/2022] Open
Abstract
PROBLEM A strong association between periodontitis and higher susceptibility to pregnancy complications like preeclampsia has been reported although the mechanisms remain elusive. Trophoblast cells modulate the recruitment and functional shaping of maternal leukocytes at early stages to sustain an antiinflammatory microenvironment and fetal growth. Neutrophil activation with reactive oxygen species (ROS) release is associated with preeclampsia. Our aim was to study the effect of the gingival crevicular fluid (GCF) from pregnant women on trophoblast cell function and trophoblast-neutrophil interaction. METHOD OF STUDY Pregnant women at 16-20 weeks of gestation (n = 27) and non-pregnant women (n = 8) as the control group were studied for gingivoperiodontal clinical score evaluation and GCF collection. Total bacteria and common periodontal pathogens were analyzed in GCF samples. The effect of each GCF sample was tested on first trimester trophoblast-derived cells to assess cell migration, cytokine expression and glucose uptake. Also, the effect of GCF on human peripheral neutrophil chemoattraction by trophoblast cells and ROS formation was assessed. RESULTS Gingival crevicular fluid from pregnant women reduced trophoblast cell migration, increased proinflammatory marker expression and glucose uptake. A significant correlation between gingivoperiodontal score and trophoblast dysfunction was observed. Upon conditioning of trophoblast cells with GCF, only the GCF from pregnant women stimulated neutrophil chemoattraction. Similarly, GCF from pregnant but not from non-pregnant controls stimulated ROS formation in neutrophils. CONCLUSIONS Gingival crevicular fluid from pregnant women is deleterious for first trimester trophoblast cell function. These effects could lead to placental homeostasis disruption underlying a pathogenic mechanism of pregnancy complications associated to periodontal disease.
Collapse
Affiliation(s)
- Vanesa Hauk
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Laboratorio de Inmunofarmacología, Buenos Aires, Argentina
| | - Luciana D'Eramo
- Universidad de Buenos Aires - Facultad de Odontología, Cátedra de Odontología Preventiva y Comunitaria, Buenos Aires, Argentina
| | - Guillermina Calo
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Laboratorio de Inmunofarmacología, Buenos Aires, Argentina
| | - Fátima Merech
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Laboratorio de Inmunofarmacología, Buenos Aires, Argentina
| | - Luciana Doga
- Universidad de Buenos Aires - Facultad de Odontología, Cátedra de Odontología Preventiva y Comunitaria, Buenos Aires, Argentina
| | - Brenda Lara
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Laboratorio de Inmunofarmacología, Buenos Aires, Argentina
| | - Laura Gliosca
- Universidad de Buenos Aires - Facultad de Odontología, Cátedra de Microbiología, Buenos Aires, Argentina
| | - Carla Massone
- Universidad de Buenos Aires - Facultad de Odontología, Cátedra de Microbiología, Buenos Aires, Argentina
| | - Susana Molgatini
- Universidad de Buenos Aires - Facultad de Odontología, Cátedra de Microbiología, Buenos Aires, Argentina
| | - Rosanna Ramhorst
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Laboratorio de Inmunofarmacología, Buenos Aires, Argentina
| | - Aldo Squassi
- Universidad de Buenos Aires - Facultad de Odontología, Cátedra de Odontología Preventiva y Comunitaria, Buenos Aires, Argentina
| | - Claudia Pérez Leirós
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Laboratorio de Inmunofarmacología, Buenos Aires, Argentina
| |
Collapse
|
30
|
Arenas-Hernandez M, Romero R, Gershater M, Tao L, Xu Y, Garcia-Flores V, Pusod E, Miller D, Galaz J, Motomura K, Schwenkel G, Para R, Gomez-Lopez N. Specific innate immune cells uptake fetal antigen and display homeostatic phenotypes in the maternal circulation. J Leukoc Biol 2022; 111:519-538. [PMID: 34889468 PMCID: PMC8881318 DOI: 10.1002/jlb.5hi0321-179rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 10/25/2021] [Accepted: 10/29/2021] [Indexed: 12/22/2022] Open
Abstract
Pregnancy represents a period when the mother undergoes significant immunological changes to promote tolerance of the fetal semi-allograft. Such tolerance results from the exposure of the maternal immune system to fetal antigens (Ags), a process that has been widely investigated at the maternal-fetal interface and in the adjacent draining lymph nodes. However, the peripheral mechanisms of maternal-fetal crosstalk are poorly understood. Herein, we hypothesized that specific innate immune cells interact with fetal Ags in the maternal circulation. To test this hypothesis, a mouse model was utilized in which transgenic male mice expressing the chicken ovalbumin (OVA) Ag under the beta-actin promoter were allogeneically mated with wild-type females to allow for tracking of the fetal Ag. Fetal Ag-carrying Ly6G+ and F4/80+ cells were identified in the maternal circulation, where they were more abundant in the second half of pregnancy. Such innate immune cells displayed unique phenotypes: while Ly6G+ cells expressed high levels of MHC-II and CD80 together with low levels of pro-inflammatory cytokines, F4/80+ cells up-regulated the expression of CD86 as well as the anti-inflammatory cytokines IL-10 and TGF-β. In vitro studies using allogeneic GFP+ placental particles revealed that maternal peripheral Ly6G+ and F4/80+ cells phagocytose fetal Ags in mid and late murine pregnancy. Importantly, cytotrophoblast-derived particles were also engulfed in vitro by CD15+ and CD14+ cells from women in the second and third trimester, providing translational evidence that this process also occurs in humans. Collectively, this study demonstrates novel interactions between specific maternal circulating innate immune cells and fetal Ags, thereby shedding light on the systemic mechanisms of maternal-fetal crosstalk.
Collapse
Affiliation(s)
- Marcia Arenas-Hernandez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
- Detroit Medical Center, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Florida International University, Miami, FL, USA
| | - Meyer Gershater
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Li Tao
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yi Xu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Errile Pusod
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Derek Miller
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jose Galaz
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kenichiro Motomura
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - George Schwenkel
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Robert Para
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
31
|
Sex, Allergic Diseases and Omalizumab. Biomedicines 2022; 10:biomedicines10020328. [PMID: 35203537 PMCID: PMC8869622 DOI: 10.3390/biomedicines10020328] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 11/17/2022] Open
Abstract
Gender differences are increasingly emerging in every area of medicine including drug therapy; however, specific gender-targeted studies are infrequent. Sex is a fundamental variable, which cannot be neglected. When optimizing therapies, gender pharmacology must always be considered in order to improve the effectiveness and safety of the use of drugs. Knowledge of gender differences promotes appropriate use of therapies and greater health protection for both genders. Further development of gender research would make it possible to report on differences in the assimilation and response of the female organism as compared to the male, in order to identify potential risks and benefits that can be found between genders. Furthermore, a better understanding of sex/gender-related influences, with regard to pharmacological activity, would allow the development of personalized “tailor-made” medicines. Here, we summarize the state of knowledge on the role of sex in several allergic diseases and their treatment with omalizumab, the first biologic drug authorized for use in the field of allergology.
Collapse
|
32
|
Giaglis S, Sur Chowdhury C, van Breda SV, Stoikou M, Tiaden AN, Daoudlarian D, Schaefer G, Buser A, Walker UA, Lapaire O, Hoesli I, Hasler P, Hahn S. Circulatory Neutrophils Exhibit Enhanced Neutrophil Extracellular Trap Formation in Early Puerperium: NETs at the Nexus of Thrombosis and Immunity. Int J Mol Sci 2021; 22:ijms222413646. [PMID: 34948443 PMCID: PMC8704360 DOI: 10.3390/ijms222413646] [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: 10/25/2021] [Revised: 12/17/2021] [Accepted: 12/17/2021] [Indexed: 11/16/2022] Open
Abstract
Pregnancy is associated with elevated maternal levels of cell-free DNA of neutrophil extracellular trap (NET) origin, as circulatory neutrophils exhibit increased spontaneous NET formation, mainly driven by G-CSF and finely modulated by sex hormones. The postpartum period, on the other hand, involves physiological alterations consistent with the need for protection against infections and fatal haemorrhage. Our findings indicate that all relevant serum markers of neutrophil degranulation and NET release are substantially augmented postpartum. Neutrophil pro-NETotic activity in vitro is also upregulated particularly in post-delivery neutrophils. Moreover, maternal puerperal neutrophils exhibit a strong pro-NETotic phenotype, associated with increased levels of all key players in the generation of NETs, namely citH3, MPO, NE, and ROS, compared to non-pregnant and pregnant controls. Intriguingly, post-delivery NET formation is independent of G-CSF in contrast to late gestation and complemented by the presence of TF on the NETs, alterations in the platelet activity status, and activation of the coagulation cascade, triggered by circulating microparticles. Taken together, our results reveal the highly pro-NETotic and potentially procoagulant nature of postpartum neutrophils, bridging an overt immune activation with possible harmful thrombotic incidence.
Collapse
Affiliation(s)
- Stavros Giaglis
- Laboratory for Prenatal Medicine, Department of Biomedicine, University of Basel, 4001 Basel, Switzerland; (C.S.C.); (S.V.v.B.); (M.S.); (G.S.); (O.L.); (I.H.); (S.H.)
- University Women’s Hospital, University Hospital Basel, 4056 Basel, Switzerland
- Division of Rheumatology, Department of Internal Medicine, Kantonsspital Aarau, 5001 Aarau, Switzerland;
- Laboratory for Experimental Rheumatology, Department of Biomedicine, University of Basel, 4001 Basel, Switzerland; (A.N.T.); (D.D.); (U.A.W.)
- Department of Rheumatology, University Hospital Basel, 4031 Basel, Switzerland
- Correspondence:
| | - Chanchal Sur Chowdhury
- Laboratory for Prenatal Medicine, Department of Biomedicine, University of Basel, 4001 Basel, Switzerland; (C.S.C.); (S.V.v.B.); (M.S.); (G.S.); (O.L.); (I.H.); (S.H.)
- University Women’s Hospital, University Hospital Basel, 4056 Basel, Switzerland
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shane Vontelin van Breda
- Laboratory for Prenatal Medicine, Department of Biomedicine, University of Basel, 4001 Basel, Switzerland; (C.S.C.); (S.V.v.B.); (M.S.); (G.S.); (O.L.); (I.H.); (S.H.)
- University Women’s Hospital, University Hospital Basel, 4056 Basel, Switzerland
- Division of Rheumatology, Department of Internal Medicine, Kantonsspital Aarau, 5001 Aarau, Switzerland;
| | - Maria Stoikou
- Laboratory for Prenatal Medicine, Department of Biomedicine, University of Basel, 4001 Basel, Switzerland; (C.S.C.); (S.V.v.B.); (M.S.); (G.S.); (O.L.); (I.H.); (S.H.)
- University Women’s Hospital, University Hospital Basel, 4056 Basel, Switzerland
| | - André N. Tiaden
- Laboratory for Experimental Rheumatology, Department of Biomedicine, University of Basel, 4001 Basel, Switzerland; (A.N.T.); (D.D.); (U.A.W.)
- Department of Rheumatology, University Hospital Basel, 4031 Basel, Switzerland
| | - Douglas Daoudlarian
- Laboratory for Experimental Rheumatology, Department of Biomedicine, University of Basel, 4001 Basel, Switzerland; (A.N.T.); (D.D.); (U.A.W.)
- Department of Rheumatology, University Hospital Basel, 4031 Basel, Switzerland
| | - Guenther Schaefer
- Laboratory for Prenatal Medicine, Department of Biomedicine, University of Basel, 4001 Basel, Switzerland; (C.S.C.); (S.V.v.B.); (M.S.); (G.S.); (O.L.); (I.H.); (S.H.)
- University Women’s Hospital, University Hospital Basel, 4056 Basel, Switzerland
| | - Andreas Buser
- Swiss Red Cross, Blood Transfusion Center, Department of Internal Medicine, Division of Hematology, University Hospital Basel, 4031 Basel, Switzerland;
| | - Ulrich A. Walker
- Laboratory for Experimental Rheumatology, Department of Biomedicine, University of Basel, 4001 Basel, Switzerland; (A.N.T.); (D.D.); (U.A.W.)
- Department of Rheumatology, University Hospital Basel, 4031 Basel, Switzerland
| | - Olav Lapaire
- Laboratory for Prenatal Medicine, Department of Biomedicine, University of Basel, 4001 Basel, Switzerland; (C.S.C.); (S.V.v.B.); (M.S.); (G.S.); (O.L.); (I.H.); (S.H.)
- University Women’s Hospital, University Hospital Basel, 4056 Basel, Switzerland
| | - Irene Hoesli
- Laboratory for Prenatal Medicine, Department of Biomedicine, University of Basel, 4001 Basel, Switzerland; (C.S.C.); (S.V.v.B.); (M.S.); (G.S.); (O.L.); (I.H.); (S.H.)
- University Women’s Hospital, University Hospital Basel, 4056 Basel, Switzerland
| | - Paul Hasler
- Division of Rheumatology, Department of Internal Medicine, Kantonsspital Aarau, 5001 Aarau, Switzerland;
| | - Sinuhe Hahn
- Laboratory for Prenatal Medicine, Department of Biomedicine, University of Basel, 4001 Basel, Switzerland; (C.S.C.); (S.V.v.B.); (M.S.); (G.S.); (O.L.); (I.H.); (S.H.)
- University Women’s Hospital, University Hospital Basel, 4056 Basel, Switzerland
| |
Collapse
|
33
|
Bert S, Ward EJ, Nadkarni S. Neutrophils in pregnancy: New insights into innate and adaptive immune regulation. Immunology 2021; 164:665-676. [PMID: 34287859 PMCID: PMC8561097 DOI: 10.1111/imm.13392] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 12/16/2022] Open
Abstract
The immunology of pregnancy has been the focus of many studies to better understand how the mother is able to tolerate the presence of a semi-allogeneic fetus. Far from the initial view of pregnancy as a state of immunosuppression, successful fetal development from implantation to birth is now known to be under the control of an intricate balance of immune cells. The balance between pro-inflammatory functions used to promote embryo implantation and placental development and immunosuppressive activity to maintain maternal tolerance of the fetus is an immunological phenotype unique to pregnancy, which is dependent on the time of gestation. Neutrophils are one of a host of innate immune cells detected at the maternal-fetal interface, but very little is known of their function. In this review, we explore the emerging functions of neutrophils during pregnancy and their interactions with and regulation of T cells, a key adaptive immune cell population essential for the establishment of fetal-maternal tolerance.
Collapse
Affiliation(s)
- Serena Bert
- William Harvey Research InstituteBarts and the London School of MedicineQueen Mary UniversityLondonUK
| | - Eleanor J. Ward
- William Harvey Research InstituteBarts and the London School of MedicineQueen Mary UniversityLondonUK
| | - Suchita Nadkarni
- William Harvey Research InstituteBarts and the London School of MedicineQueen Mary UniversityLondonUK
| |
Collapse
|
34
|
Balan P, Chong YS, Lin Q, Lim TK, Li H, Wong ML, Lopez V, He HG, Seneviratne CJ. Isobaric tags for relative and absolute quantification (iTRAQ)-based quantitative analysis of the salivary proteome during healthy pregnancy and pregnancy gingivitis. J Clin Periodontol 2021; 48:1559-1569. [PMID: 34605060 DOI: 10.1111/jcpe.13559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 09/25/2021] [Indexed: 02/06/2023]
Abstract
AIM The present study aimed to investigate the salivary proteome profiles of pregnant women with gingivitis (PG) or without gingivitis (HP) and non-pregnant healthy controls (HC) by employing iTRAQ-based proteomics. MATERIALS AND METHODS Saliva samples were collected from 30 Chinese women comprising 10 subjects in each of the three groups (PG, HP, and HC). The samples were subjected to iTRAQ-based proteomics analysis, and ELISA was performed to validate the results. The subsequent observations were validated in a cohort of 48 subjects. RESULTS Pathways associated with neutrophil-mediated immune response and antioxidant defence mechanism were significantly higher in PG than HC. The abundance of salivary cystatins (S, SA, and SN) and antimicrobials were significantly decreased in PG and HP, while cystatin C and D were additionally decreased in PG. Cystatin C was mapped to all the major catabolic pathways and was the most re-wired protein in pregnancy gingivitis. Further validation demonstrated cystatin C to be significantly lower in PG than HC. CONCLUSIONS While the decrease in levels of salivary cystatins and antimicrobial proteins may predispose healthy pregnant women to pregnancy gingivitis, it may cause persistence of inflammation in pregnant women with gingivitis.
Collapse
Affiliation(s)
- Preethi Balan
- Singapore Oral Microbiomics Initiative, National Dental Research Institute Singapore, National Dental Center, Singapore.,Oral Health Academic Clinical Program, Duke NUS Medical School, Singapore
| | - Yap S Chong
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University Hospital, Singapore
| | - Qingsong Lin
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore
| | - Teck Kwang Lim
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore
| | - Huihua Li
- Oral Health Academic Clinical Program, Duke NUS Medical School, Singapore
| | - Mun L Wong
- Discipline of Primary Dental Care and Population Oral Health, Faculty of Dentistry, National University of Singapore, Singapore
| | - Violeta Lopez
- School of Nursing, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Hong Gu He
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chaminda Jayampath Seneviratne
- Singapore Oral Microbiomics Initiative, National Dental Research Institute Singapore, National Dental Center, Singapore.,Oral Health Academic Clinical Program, Duke NUS Medical School, Singapore
| |
Collapse
|
35
|
Lee KO, Kwon I, Nam HS, Park Y, Kim J, Shim Y, Erdenebileg Z, Cha MJ, Choi HJ, Choi HY, Song JW, Heo JH. Effect of leukopenia induced by cyclophosphamide on the initial stage of arterial thrombosis in mice. Thromb Res 2021; 206:111-119. [PMID: 34455128 DOI: 10.1016/j.thromres.2021.08.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/24/2021] [Accepted: 08/18/2021] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Leukocytes are found in organizing thrombi and are associated with thrombus growth. However, their role in the initial stage of thrombus formation is not well known. We investigated the role of leukocytes in the early stage of arterial thrombosis by inducing leukopenia. METHODS In this double-blind, randomized, placebo-controlled study, 72 Institute of Cancer Research mice were randomly treated with intraperitoneal 100 mg/kg cyclophosphamide or normal saline. The primary outcome was time to occlusion after FeCl3 treatment. We also compared thrombus size, histological composition, and association with peripheral blood cell counts between cyclophosphamide and control groups. RESULTS Cyclophosphamide treatment significantly decreased leukocyte counts by 82.8% compared to placebo (P < 0.001). The time to occlusion was significantly longer in the cyclophosphamide group (3.31 ± 1.59 min) than in the control group (2.30 ± 1.14 min; P = 0.003). The immunoreactivity for Ly6G-positive cells, intracellular histone H3, and released histone H3 in thrombi was significantly reduced in the cyclophosphamide group by 92.8%, 50.2%, and 34.3%, respectively. Time to occlusion had a moderate negative correlation with leukocyte count in peripheral blood (r = -0.326, P = 0.022) in the entire group. CONCLUSIONS Cyclophosphamide-induced leukopenia attenuated thrombus formation during the early stage of arterial thrombosis. Our findings suggest the potential role of leukocytes in the initial stage of arterial thrombosis.
Collapse
Affiliation(s)
- Kee Ook Lee
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea; Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Il Kwon
- Integrative Research Center for Cerebrovascular and Cardiovascular diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyo Suk Nam
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea; Integrative Research Center for Cerebrovascular and Cardiovascular diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Youngseon Park
- Integrative Research Center for Cerebrovascular and Cardiovascular diseases, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Neurology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Republic of Korea
| | - Jayoung Kim
- Integrative Research Center for Cerebrovascular and Cardiovascular diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yeseul Shim
- Integrative Research Center for Cerebrovascular and Cardiovascular diseases, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Neurology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Republic of Korea
| | - Zolzaya Erdenebileg
- Integrative Research Center for Cerebrovascular and Cardiovascular diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Myoung Jin Cha
- Department of Neurology, National Police Hospital, Seoul, Republic of Korea
| | - Hyun-Jung Choi
- Integrative Research Center for Cerebrovascular and Cardiovascular diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye-Yeon Choi
- Department of Neurology, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Jae-Woo Song
- Department of Laboratory Medicine, Yonsei University College of Medicine, Severance Children's Hospital, Seoul, Republic of Korea
| | - Ji Hoe Heo
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea; Integrative Research Center for Cerebrovascular and Cardiovascular diseases, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Neurology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Republic of Korea.
| |
Collapse
|
36
|
Cahilog Z, Zhao H, Wu L, Alam A, Eguchi S, Weng H, Ma D. The Role of Neutrophil NETosis in Organ Injury: Novel Inflammatory Cell Death Mechanisms. Inflammation 2021; 43:2021-2032. [PMID: 32830308 PMCID: PMC7443373 DOI: 10.1007/s10753-020-01294-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
NETosis is a type of regulated cell death dependent on the formation of neutrophil extracellular traps (NET), where net-like structures of decondensed chromatin and proteases are produced by polymorphonuclear (PMN) granulocytes. These structures immobilise pathogens and restrict them with antimicrobial molecules, thus preventing their spread. Whilst NETs possess a fundamental anti-microbial function within the innate immune system under physiological circumstances, increasing evidence also indicates that NETosis occurs in the pathogenic process of other disease type, including but not limited to atherosclerosis, airway inflammation, Alzheimer’s and stroke. Here, we reviewed the role of NETosis in the development of organ injury, including injury to the brain, lung, heart, kidney, musculoskeletal system, gut and reproductive system, whilst therapeutic agents in blocking injuries induced by NETosis in its primitive stages were also discussed. This review provides novel insights into the involvement of NETosis in different organ injuries, and whilst potential therapeutic measures targeting NETosis remain a largely unexplored area, these warrant further investigation.
Collapse
Affiliation(s)
- Zhen Cahilog
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, 369 Fulham Road, London, SW10 9NH, UK
| | - Hailin Zhao
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, 369 Fulham Road, London, SW10 9NH, UK
| | - Lingzhi Wu
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, 369 Fulham Road, London, SW10 9NH, UK
| | - Azeem Alam
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, 369 Fulham Road, London, SW10 9NH, UK
| | - Shiori Eguchi
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, 369 Fulham Road, London, SW10 9NH, UK
| | - Hao Weng
- Department of Anesthesiology, Shanghai Fengxian District Central Hospital, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Fengxian District, Shanghai, China
| | - Daqing Ma
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, 369 Fulham Road, London, SW10 9NH, UK.
| |
Collapse
|
37
|
Makatsariya AD, Slukhanchuk EV, Bitsadze VO, Khizroeva JK, Tretyakova MV, Makatsariya NA, Akinshina SV, Shkoda AS, Pankratyeva LL, Di Renzo GC, Rizzo G, Grigorieva KN, Tsibizova VI, Gris JC, Elalamy I. Neutrophil extracellular traps: a role in inflammation and dysregulated hemostasis as well as in patients with COVID-19 and severe obstetric pathology. OBSTETRICS, GYNECOLOGY AND REPRODUCTION 2021; 15:335-350. [DOI: 10.17749/2313-7347/ob.gyn.rep.2021.238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Numerous studies have proven a close relationship between inflammatory diseases and the state of hypercoagulability. In fact, thromboembolic complications represent one of the main causes of disability and mortality in acute and chronic inflammatory diseases, cancer and obstetric complications. Despite this, the processes of hemostasis and immune responses have long been considered separately; currently, work is underway to identify the molecular basis for a relationship between such systems. It has been identified that various pro-inflammatory stimuli are capable of triggering a coagulation cascade, which in turn modulates inflammatory responses. Neutrophil extracellular traps (NETs) are the networks of histones of extracellular DNA generated by neutrophils in response to inflammatory stimuli. The hemostasis is activated against infection in order to minimize the spread of infection and, if possible, inactivate the infectious agent. Another molecular network is based on fibrin. Over the last 10 years, there has been accumulated a whole body of evidence that NETs and fibrin are able to form a united network within a thrombus, stabilizing each other. Similarities and molecular cross-reactions are also present in the processes of fibrinolysis and lysis of NETs. Both NETs and von Willebrand factor (vWF) are involved in thrombosis as well as inflammation. During the development of these conditions, a series of events occurs in the microvascular network, including endothelial activation, NETs formation, vWF secretion, adhesion, aggregation, and activation of blood cells. The activity of vWF multimers is regulated by the specific metalloproteinase ADAMTS-13 (a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13). Studies have shown that interactions between NETs and vWF can lead to arterial and venous thrombosis and inflammation. In addition, the contents released from activated neutrophils or NETs result in decreased ADAMTS-13 activity, which can occur in both thrombotic microangiopathies and acute ischemic stroke. Recently, NETs have been envisioned as a cause of endothelial damage and immunothrombosis in COVID-19. In addition, vWF and ADAMTS-13 levels predict COVID-19 mortality. In this review, we summarize the biological characteristics and interactions of NETs, vWF, and ADAMTS-13, the effect of NETs on hemostasis regulation and discuss their role in thrombotic conditions, sepsis, COVID-19, and obstetric complications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - A. S. Shkoda
- Vorokhobov City Clinical Hospital № 67, Moscow Healthcare Department
| | - L. L. Pankratyeva
- Vorokhobov City Clinical Hospital № 67, Moscow Healthcare Department; Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Health Ministry of Russian Federation
| | - G. C. Di Renzo
- Sechenov University; Center for Prenatal and Reproductive Medicine, University of Perugia
| | - G. Rizzo
- Sechenov University; University of Rome Tor Vergata
| | | | - V. I. Tsibizova
- Almazov National Medical Research Centre, Health Ministry of Russian Federation
| | - J.-C. Gris
- Sechenov University; University of Montpellier
| | - I. Elalamy
- Sechenov University; Medicine Sorbonne University; Hospital Tenon
| |
Collapse
|
38
|
Rodriguez D, Jerjes-Sanchez C, Fonseca S, Garcia-Toto R, Martinez-Alvarado J, Panneflek J, Ortiz-Ledesma C, Nevarez F. Thrombolysis in massive and submassive pulmonary embolism during pregnancy and the puerperium: a systematic review. J Thromb Thrombolysis 2021; 50:929-941. [PMID: 32347509 DOI: 10.1007/s11239-020-02122-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Thrombolysis in high-risk pulmonary embolism (PE) patients is recommended worldwide; however, the evidence for thrombolysis during pregnancy and the immediate puerperium remains unclear. We conducted a systematic review from 1950 to 2019 through PubMed, Ovid/Willey, and Cochrane Library to assess the safety and effectiveness of thrombolysis during pregnancy and the immediate puerperium. Additionally, we characterized the clinical presentation, risk stratification, and diagnostic approach. We have communicated our results according to the PRISMA statement. We collected 141 records and, after critical assessment, included 47 case reports of 54 patients, including 43 and 11 patients during pregnancy and puerperium, respectively. During pregnancy, alteplase was the most frequent systemic thrombolytic agent used (67%), but only nine patients received the approved FDA regimen. With catheter-directed thrombolysis, low-dose thrombolytics and fragmentation were the most common regimens. Major bleeding occurred in 18% of cases, but there was no intracranial bleeding. One maternal death occurred secondary to refractory cardiogenic shock. Fetal mortality was 20%. During the immediate puerperium, nine patients received "off-label" first-, second-, and third-generation thrombolytic regimens, and four cases underwent catheter-directed thrombolysis. We observed nine major bleeding events, seven of which were from the uterine location and none of which were intracranial. In conclusion, overall, these data do not suggest prohibitive risk associated with thrombolysis for PE in pregnancy. Management of massive and high-risk submassive PE in pregnancy should be individualized to each patient. In the data presented, no fatal bleeding or intracranial bleeding was observed. Finally, future efforts should systematically collect and report data on high-risk PE in pregnancy and peripartum patients to improve the evidence-base clinical practice.
Collapse
Affiliation(s)
- David Rodriguez
- Escuela de Medicina y Ciencias de La Salud, Tecnologico de Monterrey, Monterrey, Nuevo Leon, Mexico
- Centro de Investigacion Biomedica del Hospital Zambrano Hellion, TecSalud, San Pedro Garza García, Nuevo Leon, Mexico
- Instituto de Cardiología y Medicina Vascular, TecSalud, San Pedro Garza García, Nuevo Leon, Mexico
| | - Carlos Jerjes-Sanchez
- Escuela de Medicina y Ciencias de La Salud, Tecnologico de Monterrey, Monterrey, Nuevo Leon, Mexico.
- Centro de Investigacion Biomedica del Hospital Zambrano Hellion, TecSalud, San Pedro Garza García, Nuevo Leon, Mexico.
- Instituto de Cardiología y Medicina Vascular, TecSalud, San Pedro Garza García, Nuevo Leon, Mexico.
- Hospital Zambrano Hellion, Batallón de San Patricio 112, Real San Agustin, San Pedro Garza Garcia, Nuevo Leon, 66278, Mexico.
| | - Sugely Fonseca
- Internal Medicine, Hospital San José, TecSalud, Nuevo Leon, Monterrey, Mexico
| | | | | | | | - Claudia Ortiz-Ledesma
- Escuela de Medicina y Ciencias de La Salud, Tecnologico de Monterrey, Monterrey, Nuevo Leon, Mexico
| | - Francisco Nevarez
- Escuela de Medicina y Ciencias de La Salud, Tecnologico de Monterrey, Monterrey, Nuevo Leon, Mexico
| |
Collapse
|
39
|
Fontoura MA, Rocha RF, Marques RE. Neutrophil Recruitment and Participation in Severe Diseases Caused by Flavivirus Infection. Life (Basel) 2021; 11:717. [PMID: 34357089 PMCID: PMC8304117 DOI: 10.3390/life11070717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/15/2021] [Accepted: 07/19/2021] [Indexed: 12/21/2022] Open
Abstract
Neutrophils are first-line responders to infections and are recruited to target tissues through the action of chemoattractant molecules, such as chemokines. Neutrophils are crucial for the control of bacterial and fungal infections, but their role in the context of viral infections has been understudied. Flaviviruses are important human viral pathogens transmitted by arthropods. Infection with a flavivirus may result in a variety of complex disease manifestations, including hemorrhagic fever, encephalitis or congenital malformations. Our understanding of flaviviral diseases is incomplete, and so is the role of neutrophils in such diseases. Here we present a comprehensive overview on the participation of neutrophils in severe disease forms evolving from flavivirus infection, focusing on the role of chemokines and their receptors as main drivers of neutrophil function. Neutrophil activation during viral infection was shown to interfere in viral replication through effector functions, but the resulting inflammation is significant and may be detrimental to the host. For congenital infections in humans, neutrophil recruitment mediated by CXCL8 would be catastrophic. Evidence suggests that control of neutrophil recruitment to flavivirus-infected tissues may reduce immunopathology in experimental models and patients, with minimal loss to viral clearance. Further investigation on the roles of neutrophils in flaviviral infections may reveal unappreciated functions of this leukocyte population while increasing our understanding of flaviviral disease pathogenesis in its multiple forms.
Collapse
Affiliation(s)
- Marina Alves Fontoura
- Brazilian Biosciences National Laboratory—LNBio, Brazilian Center for Research in Energy and Materials—CNPEM, Campinas 13083-100, Brazil; (M.A.F.); (R.F.R.)
- Cellular and Structural Biology Graduate Program, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083-865, Brazil
| | - Rebeca Fróes Rocha
- Brazilian Biosciences National Laboratory—LNBio, Brazilian Center for Research in Energy and Materials—CNPEM, Campinas 13083-100, Brazil; (M.A.F.); (R.F.R.)
- Genetics and Molecular Biology Graduate Program, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083-970, Brazil
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Rafael Elias Marques
- Brazilian Biosciences National Laboratory—LNBio, Brazilian Center for Research in Energy and Materials—CNPEM, Campinas 13083-100, Brazil; (M.A.F.); (R.F.R.)
| |
Collapse
|
40
|
Al-Ofi E, Alrafiah A, Maidi S, Almaghrabi S, Hakami N. Altered Expression of Angiogenic Biomarkers in Pregnancy Associated with Gestational Diabetes. Int J Gen Med 2021; 14:3367-3375. [PMID: 34285561 PMCID: PMC8286247 DOI: 10.2147/ijgm.s316670] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/29/2021] [Indexed: 12/26/2022] Open
Abstract
Background Gestational diabetes mellitus (GDM) typically occurs during the third trimester of pregnancy. Maternal hyperglycemic may influence the expression of pro-and anti-angiogenic factors. Altered levels of angiogenic biomarkers in GDM pregnant women are associated with abnormal placentation. This study aimed to investigate the rates of expression of five angiogenic biomarkers called vascular endothelial growth factor-A (VEGF-A), angiopoietin-2, endoglin, endothelin-1, and granulocyte colony-stimulating factor (G-CSF) in GDM. Methods The samples were obtained from normal (n=9) and GDM (n=10) pregnancies. Multiplex assay was used to assess the levels of angiogenic biomarkers including VEGF-A, endoglin, endothelin-1, angiopoietin-2, and G-CSF in serum samples. All data were statistically analyzed using an unpaired Student’s t-test. Correlations between measured parameters were made using Pearson correlations. Results VEGF-A, endoglin, endothelin-1, and angiopoietin-2 levels in GDM were significantly higher (P value = 0.001, 0.042, 0.049, 0.001; respectively) compared to control. However, G-CSF level exhibited a non-significant increase (P=0.466) in GDM compared to healthy controls. There was a significant positive correlation between angiopoietin-2 with endoglin, endothelin-1, and VEGF-A. Moreover, there was a significant positive correlation between VEGF-A with endoglin and endothelin-1. Most interestingly, there was a significant positive correlation between G-CSF with endothelin-1. Conclusion The angiogenic biomarkers were highly altered in pregnant women with GDM. The study provides a novel advance in the field of gestational diabetes, in terms of increase of angiogenic factors that can modify the vascularization of the placenta, the development of fetal vascular system and the insulin resistance itself.
Collapse
Affiliation(s)
- Ebtisam Al-Ofi
- Department of Physiology, Faculty of Medicine, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Aziza Alrafiah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Salman Maidi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Safa Almaghrabi
- Department of Physiology, Faculty of Medicine, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Nora Hakami
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King AbdulAziz University, Jeddah, Saudi Arabia
| |
Collapse
|
41
|
Balan P, Chong YS, Lin Q, Lim TK, Suriyanarayanan T, Udawatte NS, Wong ML, Lopez V, He HG, Seneviratne CJ. Salivary Proteomic Profiling Identifies Role of Neutrophil Extracellular Traps Formation in Pregnancy Gingivitis. Immunol Invest 2021; 51:103-119. [PMID: 33902370 DOI: 10.1080/08820139.2020.1810704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Pregnancy gingivitis peaks during mid-pregnancy and resolves transiently towards the postpartum period. However, the role of maternal immune response in orchestrating gingival inflammation has not yet been fully understood. Hence, in this study, we examined the salivary protein profile during the three trimesters of pregnancy, in context to pregnancy gingivitis, employing iTRAQ-based quantitative proteomics. Unstimulated saliva was collected from 10 subjects in each trimester of pregnancy and postpartum period. Samples were analysed using iTRAQ analysis and ELISA and SEM was performed to validate results. Neutrophil mediated immune response was overrepresented in all three trimesters of pregnancy, despite the decrease in phagocytic responses during the second and third trimesters. ELISA showed a significantly higher Neutrophil Extracellular Traps (NETs) formation in the third trimester of pregnancy coinciding with the resolution of pregnancy gingivitis. The NETs-associated proteins (neutrophil elastase and myeloperoxidase) showed a positive correlation with estrogen hormones, which was also highest during the third trimester. Sex hormone-driven NETs formation could be the mainstay of defence that contributes to the remission of pregnancy gingivitis. This study has provided a new insight into the role of immune-modulation in pregnancy gingivitis, which will aid development of new therapeutics for managing pregnancy gingivitis in future.
Collapse
Affiliation(s)
- Preethi Balan
- Singapore Oral Microbiomics Initiative, National Dental Research Institute Singapore, National Dental Center Singapore.,Oral health Academic Clinical Program, Duke NUS Medical School, Singapore
| | - Yap Seng Chong
- Department of Obstetrics and Gynecology, National University Hospital, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Qingsong Lin
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore
| | - Teck Kwang Lim
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore
| | - Tanujaa Suriyanarayanan
- Singapore Oral Microbiomics Initiative, National Dental Research Institute Singapore, National Dental Center Singapore.,Oral health Academic Clinical Program, Duke NUS Medical School, Singapore
| | - Nadeeka Shiyamalee Udawatte
- Singapore Oral Microbiomics Initiative, National Dental Research Institute Singapore, National Dental Center Singapore
| | - Mun Loke Wong
- Discipline of Oral Sciences, Faculty of Science, National University of Singapore, Singapore
| | - Violeta Lopez
- School of Nursing, Hubei University of Medicine, Shiyan, China
| | - Hong-Gu He
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chaminda Jayampath Seneviratne
- Singapore Oral Microbiomics Initiative, National Dental Research Institute Singapore, National Dental Center Singapore.,Oral health Academic Clinical Program, Duke NUS Medical School, Singapore
| |
Collapse
|
42
|
Alhussien MN, Dang AK. JAK3 and PI3K mediates the suppressive effects of interferon tau on neutrophil extracellular traps formation during peri-implantation period. J Reprod Immunol 2021; 145:103321. [PMID: 33862433 DOI: 10.1016/j.jri.2021.103321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/29/2021] [Accepted: 04/06/2021] [Indexed: 02/06/2023]
Abstract
Interferon tau (IFNτ) is the main maternal signal for pregnancy in ruminants and modulates the functions of various immune cells, including neutrophils. Neutrophil extracellular traps (NETs) are one of the main defence mechanisms of neutrophils. In this study, we observed higher (p < 0.01) ex-vivo NETs extrusion by blood neutrophils from day 16-18 post artificial insemination (AI) in non-inseminated and inseminated non-pregnant cows compared to pregnant cows. In vitro study also showed that IFNτ hampers NETs formation in dose and time dependent manner. The lowest (p < 0.01) NETs formation and the highest (p < 0.01) mRNA expression (RT-PCR) of IFNτ stimulated genes (ISG15, OAS1, MX1) were observed when neutrophil incubated with 9 ng/mL IFNτ for 3.5 h. Signalling cascades mediating IFNτ impairment of NETs formation were identified using inhibitors of JAK2, JAK3, p38, PI3K/Akt and MAPK/Erk. IFNτ reduced (p < 0.01) the mRNA expression (RT-PCR) and concentration (ELISA) of genes and proteins that mediate NETs formation in blood neutrophils including histones (H1, H2), neutrophil elastase (NE) and myeloperoxidase (MPO). However, the effects of IFNτ on these genes and proteins were eliminated in the presence of JAK3 or PI3K inhibitors. Immunocytochemistry study also showed strong MPO signal in the presence of JAK3 or PI3K inhibitors as compared to positive control (PC, IFNτ alone). The results indicate that IFNτ impairs NETs formation using JAK3 and PI3K and thus essential for successful implantation and establishment of pregnancy in cows.
Collapse
Affiliation(s)
- Mohanned Naif Alhussien
- Lactation and Immuno-Physiology Laboratory, ICAR-National Dairy Research Institute, Karnal, Haryana, 132 001, India.
| | - Ajay Kumar Dang
- Lactation and Immuno-Physiology Laboratory, ICAR-National Dairy Research Institute, Karnal, Haryana, 132 001, India.
| |
Collapse
|
43
|
Peddapalli A, Gehani M, Kalle AM, Peddapalli SR, Peter AE, Sharad S. Demystifying Excess Immune Response in COVID-19 to Reposition an Orphan Drug for Down-Regulation of NF-κB: A Systematic Review. Viruses 2021; 13:378. [PMID: 33673529 PMCID: PMC7997247 DOI: 10.3390/v13030378] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/12/2021] [Accepted: 02/23/2021] [Indexed: 12/18/2022] Open
Abstract
The immunological findings from autopsies, biopsies, and various studies in COVID-19 patients show that the major cause of morbidity and mortality in COVID-19 is excess immune response resulting in hyper-inflammation. With the objective to review various mechanisms of excess immune response in adult COVID-19 patients, Pubmed was searched for free full articles not related to therapeutics or co-morbid sub-groups, published in English until 27.10.2020, irrespective of type of article, country, or region. Joanna Briggs Institute's design-specific checklists were used to assess the risk of bias. Out of 122 records screened for eligibility, 42 articles were included in the final review. The review found that eventually, most mechanisms result in cytokine excess and up-regulation of Nuclear Factor-κB (NF-κB) signaling as a common pathway of excess immune response. Molecules blocking NF-κB or targeting downstream effectors like Tumour Necrosis Factor α (TNFα) are either undergoing clinical trials or lack specificity and cause unwanted side effects. Neutralization of upstream histamine by histamine-conjugated normal human immunoglobulin has been demonstrated to inhibit the nuclear translocation of NF-κB, thereby preventing the release of pro-inflammatory cytokines Interleukin (IL) 1β, TNF-α, and IL-6 and IL-10 in a safer manner. The authors recommend repositioning it in COVID-19.
Collapse
Affiliation(s)
- Apparao Peddapalli
- Department of Microbiology, King George Hospital, Visakhapatnam 531011, Andhra Pradesh, India;
| | - Manish Gehani
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani-Hyderabad Campus, Hyderabad 500078, Telangana, India;
| | - Arunasree M. Kalle
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, Telangana, India;
| | - Siva R. Peddapalli
- Department of Biological Sciences-Biotechnology, Florida Institute of Technology, Melbourne, FL 32901, USA;
| | - Angela E. Peter
- Department of Biotechnology, College of Science & Technology, Andhra University, Visakhapatnam 530003, Andhra Pradesh, India;
| | - Shashwat Sharad
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD 20817, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| |
Collapse
|
44
|
Shepherd R, Cheung AS, Pang K, Saffery R, Novakovic B. Sexual Dimorphism in Innate Immunity: The Role of Sex Hormones and Epigenetics. Front Immunol 2021; 11:604000. [PMID: 33584674 PMCID: PMC7873844 DOI: 10.3389/fimmu.2020.604000] [Citation(s) in RCA: 156] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/04/2020] [Indexed: 12/17/2022] Open
Abstract
Sexual dimorphism refers to differences between biological sexes that extend beyond sexual characteristics. In humans, sexual dimorphism in the immune response has been well demonstrated, with females exhibiting lower infection rates than males for a variety of bacterial, viral, and parasitic pathogens. There is also a substantially increased incidence of autoimmune disease in females compared to males. Together, these trends indicate that females have a heightened immune reactogenicity to both self and non-self-molecular patterns. However, the molecular mechanisms driving the sexually dimorphic immune response are not fully understood. The female sex hormones estrogen and progesterone, as well as the male androgens, such as testosterone, elicit direct effects on the function and inflammatory capacity of immune cells. Several studies have identified a sex-specific transcriptome and methylome, independent of the well-described phenomenon of X-chromosome inactivation, suggesting that sexual dimorphism also occurs at the epigenetic level. Moreover, distinct alterations to the transcriptome and epigenetic landscape occur in synchrony with periods of hormonal change, such as puberty, pregnancy, menopause, and exogenous hormone therapy. These changes are also mirrored by changes in immune cell function. This review will outline the evidence for sex hormones and pregnancy-associated hormones as drivers of epigenetic change, and how this may contribute to the sexual dimorphism. Determining the effects of sex hormones on innate immune function is important for understanding sexually dimorphic autoimmune diseases, sex-specific responses to pathogens and vaccines, and how innate immunity is altered during periods of hormonal change (endogenous or exogenous).
Collapse
Affiliation(s)
- Rebecca Shepherd
- Epigenetics Group, Infection and Immunity Theme, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Ada S. Cheung
- Department of Medicine (Austin Health), The University of Melbourne, Parkville, VIC, Australia
- Department of Endocrinology, Austin Health, Heidelberg, VIC, Australia
| | - Ken Pang
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Brain and Mitochondrial Research, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC, Australia
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Adolescent Medicine, Royal Children’s Hospital, Parkville, VIC, Australia
| | - Richard Saffery
- Epigenetics Group, Infection and Immunity Theme, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Boris Novakovic
- Epigenetics Group, Infection and Immunity Theme, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
45
|
Abstract
NETosis is an innate immune response occurring after infection or inflammation: activated neutrophils expel decondensed DNA in complex with histones into the extracellular environment in a controlled manner. It activates coagulation and fuels the risk of thrombosis. Human pregnancy is associated with a mild proinflammatory state characterized by circulatory neutrophil activation which is further increased in complicated pregnancies, placenta-mediated complications being associated with an increased thrombotic risk. This aberrant activation leads to an increased release of nucleosomes in the blood flow. The aim of our study was to initially quantify nucleosome-bound histones in normal pregnancy and in placenta-mediated complication counterpart. We analyzed the role of histones on extravillous trophoblast function. Circulating nucleosome-bound histones H3 (Nu.QH3.1, Nu.QH3PanCit, Nu.QH3K27me3) and H4 (Nu.QH4K16Ac) were increased in complicated pregnancies. In vitro using the extravillous cell line HTR-8/SVNeo, we observed that free recombinant H2B, H3, and H4 inhibited migration in wound healing assay, but only H3 also blocked invasion in Matrigel-coated Transwell experiments. H3 and H4 also induced apoptosis, whereas H2B did not. Finally, the negative effects of H3 on invasion and apoptosis could be restored with enoxaparin, a low-molecular-weight heparin (LMWH), but not with aspirin. Different circulating nucleosome-bound histones are increased in complicated pregnancy and this would affect migration, invasion, and induce apoptosis of extravillous trophoblasts. Histones might be part of the link between the risk of thrombosis and pregnancy complications, with an effect of LMWH on both.
Collapse
|
46
|
Jehan F, Sazawal S, Baqui AH, Nisar MI, Dhingra U, Khanam R, Ilyas M, Dutta A, Mitra DK, Mehmood U, Deb S, Mahmud A, Hotwani A, Ali SM, Rahman S, Nizar A, Ame SM, Moin MI, Muhammad S, Chauhan A, Begum N, Khan W, Das S, Ahmed S, Hasan T, Khalid J, Rizvi SJR, Juma MH, Chowdhury NH, Kabir F, Aftab F, Quaiyum A, Manu A, Yoshida S, Bahl R, Rahman A, Pervin J, Winston J, Musonda P, Stringer JSA, Litch JA, Ghaemi MS, Moufarrej MN, Contrepois K, Chen S, Stelzer IA, Stanley N, Chang AL, Hammad GB, Wong RJ, Liu C, Quaintance CC, Culos A, Espinosa C, Xenochristou M, Becker M, Fallahzadeh R, Ganio E, Tsai AS, Gaudilliere D, Tsai ES, Han X, Ando K, Tingle M, Marić I, Wise PH, Winn VD, Druzin ML, Gibbs RS, Darmstadt GL, Murray JC, Shaw GM, Stevenson DK, Snyder MP, Quake SR, Angst MS, Gaudilliere B, Aghaeepour N. Multiomics Characterization of Preterm Birth in Low- and Middle-Income Countries. JAMA Netw Open 2020; 3:e2029655. [PMID: 33337494 PMCID: PMC7749442 DOI: 10.1001/jamanetworkopen.2020.29655] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
IMPORTANCE Worldwide, preterm birth (PTB) is the single largest cause of deaths in the perinatal and neonatal period and is associated with increased morbidity in young children. The cause of PTB is multifactorial, and the development of generalizable biological models may enable early detection and guide therapeutic studies. OBJECTIVE To investigate the ability of transcriptomics and proteomics profiling of plasma and metabolomics analysis of urine to identify early biological measurements associated with PTB. DESIGN, SETTING, AND PARTICIPANTS This diagnostic/prognostic study analyzed plasma and urine samples collected from May 2014 to June 2017 from pregnant women in 5 biorepository cohorts in low- and middle-income countries (LMICs; ie, Matlab, Bangladesh; Lusaka, Zambia; Sylhet, Bangladesh; Karachi, Pakistan; and Pemba, Tanzania). These cohorts were established to study maternal and fetal outcomes and were supported by the Alliance for Maternal and Newborn Health Improvement and the Global Alliance to Prevent Prematurity and Stillbirth biorepositories. Data were analyzed from December 2018 to July 2019. EXPOSURES Blood and urine specimens that were collected early during pregnancy (median sampling time of 13.6 weeks of gestation, according to ultrasonography) were processed, stored, and shipped to the laboratories under uniform protocols. Plasma samples were assayed for targeted measurement of proteins and untargeted cell-free ribonucleic acid profiling; urine samples were assayed for metabolites. MAIN OUTCOMES AND MEASURES The PTB phenotype was defined as the delivery of a live infant before completing 37 weeks of gestation. RESULTS Of the 81 pregnant women included in this study, 39 had PTBs (48.1%) and 42 had term pregnancies (51.9%) (mean [SD] age of 24.8 [5.3] years). Univariate analysis demonstrated functional biological differences across the 5 cohorts. A cohort-adjusted machine learning algorithm was applied to each biological data set, and then a higher-level machine learning modeling combined the results into a final integrative model. The integrated model was more accurate, with an area under the receiver operating characteristic curve (AUROC) of 0.83 (95% CI, 0.72-0.91) compared with the models derived for each independent biological modality (transcriptomics AUROC, 0.73 [95% CI, 0.61-0.83]; metabolomics AUROC, 0.59 [95% CI, 0.47-0.72]; and proteomics AUROC, 0.75 [95% CI, 0.64-0.85]). Primary features associated with PTB included an inflammatory module as well as a metabolomic module measured in urine associated with the glutamine and glutamate metabolism and valine, leucine, and isoleucine biosynthesis pathways. CONCLUSIONS AND RELEVANCE This study found that, in LMICs and high PTB settings, major biological adaptations during term pregnancy follow a generalizable model and the predictive accuracy for PTB was augmented by combining various omics data sets, suggesting that PTB is a condition that manifests within multiple biological systems. These data sets, with machine learning partnerships, may be a key step in developing valuable predictive tests and intervention candidates for preventing PTB.
Collapse
Affiliation(s)
- Fyezah Jehan
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Sunil Sazawal
- Centre for Public Health Kinetics, New Delhi, Delhi, India
| | - Abdullah H. Baqui
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Muhammad Imran Nisar
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Usha Dhingra
- Centre for Public Health Kinetics, New Delhi, Delhi, India
| | - Rasheda Khanam
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Muhammad Ilyas
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Arup Dutta
- Centre for Public Health Kinetics, New Delhi, Delhi, India
| | - Dipak K. Mitra
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Usma Mehmood
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Saikat Deb
- Centre for Public Health Kinetics, New Delhi, Delhi, India
- Public Health Laboratory-Ivo de Carneri, Pemba Island, Zanzibar
| | - Arif Mahmud
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Aneeta Hotwani
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | | | - Sayedur Rahman
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Ambreen Nizar
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | | | - Mamun Ibne Moin
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Sajid Muhammad
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | | | - Nazma Begum
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Waqasuddin Khan
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Sayan Das
- Centre for Public Health Kinetics, New Delhi, Delhi, India
| | - Salahuddin Ahmed
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Tarik Hasan
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Javairia Khalid
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Syed Jafar Raza Rizvi
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | | - Nabidul Haque Chowdhury
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Furqan Kabir
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Fahad Aftab
- Centre for Public Health Kinetics, New Delhi, Delhi, India
| | - Abdul Quaiyum
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Alexander Manu
- Maternal, Newborn, Child and Adolescent Health Research, World Health Organization, Geneva, Switzerland
| | - Sachiyo Yoshida
- Maternal, Newborn, Child and Adolescent Health Research, World Health Organization, Geneva, Switzerland
| | - Rajiv Bahl
- Maternal, Newborn, Child and Adolescent Health Research, World Health Organization, Geneva, Switzerland
| | - Anisur Rahman
- Matlab Health Research Centre, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Jesmin Pervin
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Jennifer Winston
- Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill
| | - Patrick Musonda
- School of Public Health, University of Zambia, Lusaka, Zambia
| | - Jeffrey S. A. Stringer
- Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill
| | - James A. Litch
- Global Alliance to Prevent Prematurity and Stillbirth, Seattle, Washington
| | - Mohammad Sajjad Ghaemi
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
- Digital Technologies Research Centre, National Research Council Canada, Toronto, Ontario, Canada
| | - Mira N. Moufarrej
- Department of Bioengineering, Stanford University, Stanford, California
| | - Kévin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Songjie Chen
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Ina A. Stelzer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Natalie Stanley
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Alan L. Chang
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Ghaith Bany Hammad
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Ronald J. Wong
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Candace Liu
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | | | - Anthony Culos
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Camilo Espinosa
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Maria Xenochristou
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Martin Becker
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Ramin Fallahzadeh
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Edward Ganio
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Amy S. Tsai
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Dyani Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Eileen S. Tsai
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Xiaoyuan Han
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Kazuo Ando
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Martha Tingle
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Ivana Marić
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Paul H. Wise
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Virginia D. Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California
| | - Maurice L. Druzin
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California
| | - Ronald S. Gibbs
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California
| | - Gary L. Darmstadt
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | | | - Gary M. Shaw
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - David K. Stevenson
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Stephen R. Quake
- Department of Bioengineering, Stanford University, Stanford, California
| | - Martin S. Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
- Department of Biomedical Informatics, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
47
|
Makatsariya A, Slukhanchuk E, Bitsadze V, Khizroeva J, Tretyakova M, Tsibizova V, Dobryakov A, Elalamy I, Gris JC. COVID-19, neutrophil extracellular traps and vascular complications in obstetric practice. J Perinat Med 2020; 48:985-994. [PMID: 32739908 DOI: 10.1515/jpm-2020-0280] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 07/20/2020] [Indexed: 02/07/2023]
Abstract
An issue of the novel coronavirus infection spreading is currently in the first place among others in the list of the international medical community. Due to lack of information, conflicting research findings, multicomponent effect of the virus on the body host, as well as various consequences that the virus triggers in the body, now every medical specialty does study the viral attack pathogenesis. Recent months showed that vascular complications are the most severe in the Coronavirus Disease 2019 (COVID-19) and are the main cause of death in the patients. The mechanisms of vascular complications are complex and affect both the hemostatic system and immune responses, "inflammatory storm", disorders of the renin-angiotensin-aldosterone system, endotheliopathy, etc. Due to the leading role of vascular complications in the viral infection pathogenesis, several groups of patients are at extra risk, including pregnant women, patients with a burdened obstetric history, with hereditary thrombophilia and antiphospholipid syndrome, and patients after in vitro fertilization (IVF). In this category of pregnant women, use of low-molecular-weight heparins (LMWH) is particularly important for both prevention of vascular and obstetric complications, and for pathogenetic therapy of COVID-19.
Collapse
Affiliation(s)
- Alexander Makatsariya
- Department of Obstetrics and Gynecology, I.M.Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | - Viktoriya Bitsadze
- Department of Obstetrics and Gynecology, I.M.Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Jamilya Khizroeva
- Department of Obstetrics and Gynecology, I.M.Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | - Valentina Tsibizova
- Department of Functional and Ultrasound Diagnostics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Andrey Dobryakov
- Pathologo-Anatomical Department of City Clinical Hospital, Bakhrushin Brothers Hospital, Moscow, Russia
| | - Ismail Elalamy
- Department of Obstetrics and Gynecology, I.M.Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.,Hematology Department of Thrombosis Center, Tenon University Hospital, Medicine Sorbonne University, Paris, France
| | - Jean C Gris
- Department of Obstetrics and Gynecology, I.M.Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.,Hematology Department of Montpellier University, Montpellier, France
| |
Collapse
|
48
|
Bitsadze V, Khizroeva J, Elalamy I, Alexander M. Venous thrombosis risk factors in pregnant women. J Perinat Med 2020; 0:jpm-2020-0011. [PMID: 33098632 DOI: 10.1515/jpm-2020-0011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/09/2020] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Assess all risk factors of venous thromboembolism (VTE) in pregnancy and puerperium. METHODS Different guidelines for VTE prevention have been analyzed. RESULTS Various recommendations have appeared for practitioners during the last 10-15 years on the basis of the risk factors analysis in order to prevent VTE in pregnant women more effectively. Nevertheless, none of these recommendations can yet take into account all risk factors, although convenient scoring systems have emerged for risk assessment and clear recommendations on anti-thrombotic prophylaxis regimens in risk groups in recent years. CONCLUSIONS VTE is the third most common cause of death on Earth after myocardial infarctions and strokes, according to the World Health Organization. Pregnancy is a unique condition of woman, when enormous changes occur in functioning of the most important systems of homeostasis in a relatively short time. These are physiological hypercoagulation, slowing of blood flow, increase in circulating blood volume, etc. However, while being physiological, these changes increase the risks of venous thromboembolism by almost six times. In some cases, there appears an imbalance or dissociation between the functioning of natural antithrombotic systems and the activation of coagulation as a consequence of genetically or acquired determined causes (genetic thrombophilia, antiphospholipid syndrome, comorbidities, obstetric complications and other exogenous and endogenous factors). Accordingly, identification of risk factors, their systematization, and determination of VTE risks in pregnancy and puerperium is one of the most important tasks of clinical medicine. This article will review historical understanding of thrombosis in pregnant women, progress in understanding VTE risk factors in pregnant women, and available reserves in identifying new risk factors during pregnancy and puerperium in order to stratify risks more efficiently.
Collapse
Affiliation(s)
- Victoria Bitsadze
- Department of Obstetrics and Gynecology, Russian Academy of Sciences, I.M.Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Jamilya Khizroeva
- Department of Obstetrics and Gynecology, Russian Academy of Sciences, I.M.Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Ismail Elalamy
- Department Hematology and Thrombosis Center, Medicine, Sorbonne University, Paris, France
| | - Makatsariya Alexander
- Department of Obstetrics and Gynecology, Russian Academy of Sciences, I.M.Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| |
Collapse
|
49
|
Abu-Raya B, Michalski C, Sadarangani M, Lavoie PM. Maternal Immunological Adaptation During Normal Pregnancy. Front Immunol 2020; 11:575197. [PMID: 33133091 PMCID: PMC7579415 DOI: 10.3389/fimmu.2020.575197] [Citation(s) in RCA: 332] [Impact Index Per Article: 66.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/18/2020] [Indexed: 12/25/2022] Open
Abstract
The risk and severity of specific infections are increased during pregnancy due to a combination of physiological and immunological changes. Characterizing the maternal immune system during pregnancy is important to understand how the maternal immune system maintains tolerance towards the allogeneic fetus. This may also inform strategies to prevent maternal fatalities due to infections and optimize maternal vaccination to best protect the mother-fetus dyad and the infant after birth. In this review, we describe what is known about the immunological changes that occur during a normal pregnancy.
Collapse
Affiliation(s)
- Bahaa Abu-Raya
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada.,BC Children's Hospital Research Institute, Vancouver, BC, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.,Experimental Medicine Program, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Christina Michalski
- BC Children's Hospital Research Institute, Vancouver, BC, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.,Experimental Medicine Program, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Manish Sadarangani
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada.,BC Children's Hospital Research Institute, Vancouver, BC, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.,Experimental Medicine Program, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Pascal M Lavoie
- BC Children's Hospital Research Institute, Vancouver, BC, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.,Experimental Medicine Program, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
50
|
Cutolo M, Straub RH. Sex steroids and autoimmune rheumatic diseases: state of the art. Nat Rev Rheumatol 2020; 16:628-644. [PMID: 33009519 DOI: 10.1038/s41584-020-0503-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2020] [Indexed: 12/16/2022]
Abstract
In autoimmune rheumatic diseases, oestrogens can stimulate certain immune responses (including effects on B cells and innate immunity), but can also have dose-related anti-inflammatory effects on T cells, macrophages and other immune cells. By contrast, androgens and progesterone have predominantly immunosuppressive and anti-inflammatory effects. Hormone replacement therapies and oral contraception (and also pregnancy) enhance or decrease the severity of autoimmune rheumatic diseases at a genetic or epigenetic level. Serum androgen concentrations are often low in men and in women with autoimmune rheumatic diseases, suggesting that androgen-like compounds might be a promising therapeutic approach. However, androgen-to-oestrogen conversion (known as intracrinology) is enhanced in inflamed tissues, such as those present in patients with autoimmune rheumatic diseases. In addition, it is becoming evident that the gut microbiota differs between the sexes (known as the microgenderome) and leads to sex-dependent genetic and epigenetic changes in gastrointestinal inflammation, systemic immunity and, potentially, susceptibility to autoimmune or inflammatory rheumatic diseases. Future clinical research needs to focus on the therapeutic use of androgens and progestins or their downstream signalling cascades and on new oestrogenic compounds such as tissue-selective oestrogen complex to modulate altered immune responses.
Collapse
Affiliation(s)
- Maurizio Cutolo
- Research Laboratories and Academic Division of Clinical Rheumatology, Postgraduate School of Rheumatology, Department of Internal Medicine DIMI, University of Genova, IRCCS San Martino Polyclinic, Genoa, Italy.
| | - Rainer H Straub
- Laboratory of Experimental Rheumatology and Neuroendocrine Immunology, Division of Rheumatology, Department of Internal Medicine, University Hospital of Regensburg, Regensburg, Germany
| |
Collapse
|