1
|
Indra S, Chalak K, Das P, Mukhopadhyay A. Placenta a potential gateway of prenatal SARS-CoV-2 infection: A review. Eur J Obstet Gynecol Reprod Biol 2024; 303:123-131. [PMID: 39461078 DOI: 10.1016/j.ejogrb.2024.10.027] [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: 08/13/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024]
Abstract
SARS-CoV-2, the causative agent of COVID-19, can infect various tissues in the body apart from the lungs. Although placental infection remains controversial, COVID-19-associated placental abnormalities have been reported worldwide. Therefore, COVID-19 poses a significant risk for fetal distress as well. Scientists are currently debating whether such distress results from direct viral induced assault or placental damage caused by the mother's immune response. The placenta develops different histopathological lesions in response to maternal SARS-CoV-2 infection. While some studies support both theories, the transmission rate through the placenta remains low. Therefore, a more in-depth study is necessary to determine the primary cause of maternal SARS-CoV-2-induced fetal distress. This comprehensive review is aimed to shed light on the possible reasons towards fetal distress among mothers with COVID-19. This review describes the various mechanisms of viral entry along with the mechanisms by which the virus could affect the placenta. Reported cases of placental abnormalities and fetal distress symptoms have been collated to provide an overview of the current state of knowledge on vertical transmission of COVID-19.
Collapse
Affiliation(s)
- Subhashis Indra
- Department of Life Sciences, Presidency University, Kolkata 700073, India
| | - Kuheli Chalak
- Department of Life Sciences, Presidency University, Kolkata 700073, India
| | - Purbasha Das
- Department of Life Sciences, Presidency University, Kolkata 700073, India
| | | |
Collapse
|
2
|
Sun N, Zhang M, Kong J, Li J, Dong Y, Wang X, Fu L, Zhou Y, Chen Y, Li Y, Sun X, Guo R. Dysregulated T-cell homeostasis and decreased CD30 + Treg proliferating in aplastic anemia. Heliyon 2024; 10:e35775. [PMID: 39170389 PMCID: PMC11337026 DOI: 10.1016/j.heliyon.2024.e35775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
Aplastic anemia (AA) is an autoimmune hematopoietic disease mediated by autoreactive T cells leading to bone marrow failure. However, the precise role of autoreactive T cells in the development of AA is not fully understood, hindering the advancement of therapeutic and diagnostic strategies. In this study, we conducted a single-cell transcriptome analysis of CD8+ T cells, conventional CD4+ T (CD4+ Tconv) cells, and Treg cells, to elucidate the potential disruption of T cell homeostasis in patients with AA. We identified changes in CD4+ Tconv cells, including loss of homeostasis in naïve and memory cells and increased differentiation potential in T helper type 1 (TH1), T helper type 2 (TH2), and T helper type 17 (TH17) cells. Additionally, we identified naïve and memory CD8+ T cells that were enforced into an effector state. CD127 is an ideal surface marker for assessing the immune state of CD8+ T cells,as identified by flow cytometry. Abnormal expression of TNFSF8 has been observed in AA and other autoimmune diseases. Flow cytometry analysis revealed that TNFRSF8 (CD30), a receptor for TNFSF8, was predominantly present in human Treg cells. Importantly, patients with AA have a decreased CD30+ Treg subset. RNA-sequencing analysis revealed, that the CD30+ Treg cells are characterized by high proliferation and a remarkable immunosuppressive phenotype. Taken, together, we propose that abnormal TNFSF8/TNFRSF8 signaling is involved in dysfunctional T cell immunity by increasing the destruction of CD30+ Treg cells.
Collapse
Affiliation(s)
- Nannan Sun
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengmeng Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jingjing Kong
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jin Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yong Dong
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiaoqian Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liyan Fu
- Department of Laboratory Medicine, The First Clinical College of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yiwei Zhou
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yaoyao Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yingmei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xianlei Sun
- Basic Medical Research Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Rongqun Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
3
|
Choi CY, Gadhave K, Villano J, Pekosz A, Mao X, Jia H. Generation and characterization of a humanized ACE2 mouse model to study long-term impacts of SARS-CoV-2 infection. J Med Virol 2024; 96:e29349. [PMID: 38185937 PMCID: PMC10783855 DOI: 10.1002/jmv.29349] [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: 11/30/2023] [Accepted: 12/13/2023] [Indexed: 01/09/2024]
Abstract
Although the COVID-19 pandemic has officially ended, the persistent challenge of long-COVID or post-acute COVID sequelae (PASC) continues to impact societies globally, highlighting the urgent need for ongoing research into its mechanisms and therapeutic approaches. Our team has recently developed a novel humanized ACE2 mouse model (hACE2ki) designed explicitly for long-COVID/PASC research. This model exhibits human ACE2 expression in tissue and cell-specific patterns akin to mouse Ace2. When we exposed young adult hACE2ki mice (6 weeks old) to various SARS-CoV-2 lineages, including WA, Delta, and Omicron, at a dose of 5 × 105 PFU/mouse via nasal instillation, the mice demonstrated distinctive phenotypes characterized by differences in viral load in the lung, trachea, and nasal turbinate, weight loss, and changes in pro-inflammatory cytokines and immune cell profiles in bronchoalveolar lavage fluid. Notably, no mortality was observed in this age group. Further, to assess the model's relevance for long-COVID studies, we investigated tau protein pathologies, which are linked to Alzheimer's disease, in the brains of these mice post SARS-CoV-2 infection. Our findings revealed the accumulation and longitudinal propagation of tau, confirming the potential of our hACE2ki mouse model for preclinical studies of long-COVID.
Collapse
Affiliation(s)
- Chang-Yong Choi
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School, of Medicine, Baltimore, MD 21205, USA
| | - Kundlik Gadhave
- Institute for Cell Engineering, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jason Villano
- Molecular and Comparative Pathobiology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Andrew Pekosz
- Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Xiaobo Mao
- Institute for Cell Engineering, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Institute for NanoBioTechnology, Department of Material Science and Engineering, Johns Hopkins Whiting School of Engineering, Baltimore, MD 21218, USA
| | - Hongpeng Jia
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School, of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
4
|
Li Y, Cao H, Jiang Z, Yan K, Shi J, Wang S, Wang F, Wang W, Li X, Sun N, Liu L, Chen L, Chen Y, Guo R, Song Y. CCL17 acts as an antitumor chemokine in micromilieu‐driven immune skewing. Int Immunopharmacol 2023; 118:110078. [PMID: 37001380 DOI: 10.1016/j.intimp.2023.110078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/07/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023]
Abstract
BACKGROUND Chemokines are critical players in the local immune responses to tumors. CCL17 (thymus and activation-regulated chemokine, TARC) and CCL22 (macrophage-derived chemokine, MDC) can attract CCR4-bearing cells involving the immune landscape of cancer. However, their direct roles and functional states in tumors remain largely unclear. METHODS We analyzed the lymphoma-related scRNA-seq and bulk RNA-seq datasets and identified the CCL17/CCL22-CCR4 axis as the unique participant of the tumor microenvironment. Then we edited the A20 lymphoma cell line to express CCL17 and CCL22 and assessed their function using three mouse models (Balb/C mouse, Nude mouse, and NSG mouse). In addition, we retrospectively checked the relationship between the CCL17/CCL22-CCR4 axis and the survival rates of cancer patients. RESULTS The active CCL17/CCL22-CCR4 axis is a distinctive feature of the Hodgkin lymphoma microenvironment. CCR4 is widely expressed in immune cells but highly exists on the surface of NK, NKT, and Treg cells. The tumor model of Balb/C mice showed that CCL17 acts as an anti-tumor chemokine mediated by activated T cell response. In addition, the tumor model of Nude mice showed that CCL17 recruits NK cells for inhibiting lymphoma growth and enhances the NK-cDC1 interaction for resisting IL4i1-mediated immunosuppression. Interestingly, CCL17-mediated antitumor immune responses depend on lymphoid lineages but not mainly myeloid ones. Furthermore, we found CCL17/CCL22-CCR4 axis cannot be regarded as biomarkers of poor prognosis in most cancer types from the TCGA database. CONCLUSION We provided direct evidence of antitumor functions of CCL17 mediated by the recruitment of conventional T cells, NKT cells, and NK cells. Clinical survival outcomes of target gene (CCL17, CCL22, and CCR4) expression also identified that CCL17/CCL22-CCR4 axis is not a marker of poor prognosis.
Collapse
|
5
|
Alessa H, Peres LC, Ferriman E, Fry A, Whitby E. MRI features of perifibrinous deposits in the placenta due to COVID-19. BJR Case Rep 2023; 9:20220132. [PMID: 36873235 PMCID: PMC9976725 DOI: 10.1259/bjrcr.20220132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/09/2022] [Accepted: 11/14/2022] [Indexed: 01/13/2023] Open
Abstract
COVID-19 has been linked to pregnancy complications and loss (1). Infection during pregnancy is usually mild (2). The risk is highest in the third trimester with increased hospital admission rates and maternal and fetal compromise (3). Post-COVID placentitis is uncommon but the effect on the placenta and the fetus is extensive (4). We present a case correlating clinical, imaging, and pathological findings. Case Report: A 29-year-old para 2 gravida 1, with a normal fetal anomaly scan at 22 weeks gestational age (GA) contracted COVID at 24 weeks gestation. Fully recovered but reported reduced fetal movements at 27 weeks and 1 day. Imaging: US scan showed bright echoes within the brain, small lungs, and oligohydramnios. MRI showed abnormal brain signals, small lungs, and oligohydramnios but also a very abnormal placenta. Reduced and heterogeneous T2 signal and a marked reduction in the DWI signal intensity. The placental size was markedly reduced (volume 785.6 cm3 expected for GA is 5604.8-5952.4 cm3. The surface area of attachment was 3220 mm2, expected 22180.4-29293.2 mm2). Pathology: The placenta was small (fifth centile) with massive perivillous fibrin deposition and multifocal chronic deciduitis. Histology revealed placental chorionic villi showing diffuse sclerotic changes surrounded by perivillous fibrin deposition in the intervillous space. The basal plate revealed multifocal chronic deciduitis. When imaging the fetus, it is important to examine the placenta and correlate any abnormalities. The placenta is a forgotten organ and should be routinely included and assessed to allow the detection of important abnormalities.
Collapse
Affiliation(s)
- Hiba Alessa
- Reproductive and Developmental Medicine, University of Sheffield, Sheffield, United Kingdom
| | | | - Emma Ferriman
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Andrew Fry
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Elspeth Whitby
- Reproductive and Developmental Medicine, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
6
|
Liu Z, Zhai M, Zhang Q, Yang T, Wan Z, Li J, Liu X, Xu B, Du L, Chan RWS, Zhang L, Yeung WSB, Cheung KW, Chiu PCN, Wang WJ, Lee CL, Gao Y. Resolving the gene expression maps of human first-trimester chorionic villi with spatial transcriptome. Front Cell Dev Biol 2022; 10:1060298. [PMID: 36561369 PMCID: PMC9763897 DOI: 10.3389/fcell.2022.1060298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
The placenta is important for fetal development in mammals, and spatial transcriptomic profiling of placenta helps to resolve its structure and function. In this study, we described the landscape of spatial transcriptome of human placental villi obtained from two pregnant women at the first trimester using the modified Stereo-seq method applied for paraformaldehyde (PFA) fixation samples. The PFA fixation of human placenta villi was better than fresh villi embedded in optimum cutting temperature (OCT) compound, since it greatly improved tissue morphology and the specificity of RNA signals. The main cell types in chorionic villi such as syncytiotrophoblasts (SCT), villous cytotrophoblasts (VCT), fibroblasts (FB), and extravillous trophoblasts (EVT) were identified with the spatial transcriptome data, whereas the minor cell types of Hofbauer cells (HB) and endothelial cells (Endo) were spatially located by deconvolution of scRNA-seq data. We demonstrated that the Stereo-seq data of human villi could be used for sophisticated analyses such as spatial cell-communication and regulatory activity. We found that the SCT and VCT exhibited the most ligand-receptor pairs that could increase differentiation of the SCT, and that the spatial localization of specific regulons in different cell types was associated with the pathways related to hormones transport and secretion, regulation of mitotic cell cycle, and nutrient transport pathway in SCT. In EVT, regulatory pathways such as the epithelial to mesenchyme transition, epithelial development and differentiation, and extracellular matrix organization were identified. Finally, viral receptors and drug transporters were identified in villi according to the pathway analysis, which could help to explain the vertical transmission of several infectious diseases and drug metabolism efficacy. Our study provides a valuable resource for further investigation of the placenta development, physiology and pathology in a spatial context.
Collapse
Affiliation(s)
| | | | - Qingqing Zhang
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China,Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Tingyu Yang
- BGI-Shenzhen, Shenzhen, China,Shenzhen Engineering Laboratory for Birth Defects Screening, Shenzhen, China,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | | | - Jianlin Li
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Xiaofeng Liu
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Bo Xu
- Department of Obstetrics and Gynaecology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Libei Du
- Department of Obstetrics and Gynaecology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Rachel W. S. Chan
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China,Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Li Zhang
- Department of Obstetrics and Gynaecology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - William S. B. Yeung
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China,Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Ka Wang Cheung
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Philip C. N. Chiu
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China,Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Wen-Jing Wang
- BGI-Shenzhen, Shenzhen, China,*Correspondence: Wen-Jing Wang, ; Cheuk-Lun Lee, ; Ya Gao,
| | - Cheuk-Lun Lee
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China,Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China,*Correspondence: Wen-Jing Wang, ; Cheuk-Lun Lee, ; Ya Gao,
| | - Ya Gao
- BGI-Shenzhen, Shenzhen, China,Shenzhen Engineering Laboratory for Birth Defects Screening, Shenzhen, China,*Correspondence: Wen-Jing Wang, ; Cheuk-Lun Lee, ; Ya Gao,
| |
Collapse
|
7
|
COVID-19, Vaccination, and Female Fertility in the Czech Republic. Int J Mol Sci 2022; 23:ijms231810909. [PMID: 36142820 PMCID: PMC9501189 DOI: 10.3390/ijms231810909] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 11/03/2022] Open
Abstract
The fast-track process to approve vaccines against COVID-19 has raised questions about their safety, especially in relation to fertility. Over the last 2 years, studies have appeared monitoring female fertility, especially from assisted reproduction centers or in animal experiments. However, studies monitoring healthy populations are still limited. The aim of our study was to monitor the relevant parameters of female fertility (sex and other steroids, LH, FSH, SHBG, Antimüllerian hormone and antral follicle count) before and then 2-4 months after the third dose of vaccination against COVID-19 in a group of 25 healthy fertile woman. In addition, anti-SARS-CoV-2 and anti-SARS-CoV-2S antibodies were determined. We did not observe significant changes in the measured parameters before and after the third dose of vaccination. By comparing levels of the analytes with antibodies indicating a prior COVID-19 infection, we found that women who had experienced the disease had statistically lower levels of estrone, estradiol, SHBG and 5α-dihydroprogesterone, and conversely, higher levels of androgen active dehydroepiandrosterone and dihydrotestosterone. Our results confirm that vaccination does not affect female fertility, and that what fertile women should be worried about is not vaccination, but rather COVID-19 infection itself.
Collapse
|
8
|
Ezechukwu HC, Shi J, Fowora MA, Diya CA, Elfaki F, Adegboye OA. Fetoplacental transmission and placental response to SARS-CoV-2: Evidence from the literature. Front Med (Lausanne) 2022; 9:962937. [PMID: 36052328 PMCID: PMC9426356 DOI: 10.3389/fmed.2022.962937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/26/2022] [Indexed: 01/05/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a dreadful novel coronavirus with global health concerns among pregnant women. To date, the vertical transmission of SARS-CoV-2 during pregnancy remains controversial. We briefly report recent findings of placental response to SARS-CoV-2 infection and updates on vertical transmission. We systematically searched PubMed and Google Scholar databases according to PRISMA guidelines for studies reporting the effects of SARS-CoV-2 infection on the placenta and possibility of vertical transmission. We identified 45 studies reporting 1,280 human placentas that were analyzed by molecular pathology methods and 11,112 placenta-derived cells from a publicly available database that was analyzed using bioinformatics tools. The main finding of this study is that the SARS-CoV-2 canonical entry receptors (ACE2 and TMPRSS2) are abundantly expressed on the placenta during the first trimester, and this expression diminishes across gestational age. Out of 45 eligible studies identified, 24 (53.34%) showed no evidence of vertical transmission, 15 (33.33%) supported the hypothesis of very rare, low possibility of vertical transmission and 6 (13.33%) were indecisive and had no comment on vertical transmission. Furthermore, 433 placentas from 12 studies were also identified for placental pathology investigation. There was evidence of at least one form of maternal vascular malperfusion (MVM), 57/433 (13.1%), fetal vascular malperfusion (FVM), 81/433 (18.7%) and placental inflammation with excessive infiltration of CD3+ CD8+ lymphocytes, CD68+ macrophages and CD20+ lymphocytes in most of the eligible studies. Decidual vasculopathy (3.2%), infarction (3.2%), chronic histiocytic intervillositis (6.0%), thrombi vasculopathy (5.1%) were also observed in most of the MVM and FVM reported cases. The results indicated that SARS-CoV-2 induces placenta inflammation, and placenta susceptibility to SARS-CoV-2 decreases across the pregnancy window. Thus, SARS-CoV-2 infection in early pregnancy may adversely affect the developing fetus.
Collapse
Affiliation(s)
- Henry C. Ezechukwu
- Department of Medical Biochemistry, EKO University of Medicine and Health Sciences, Lagos, Nigeria
- School of Human Sciences, University of Western Australia, Perth, WA, Australia
| | - Jiahua Shi
- School of Medical, Indigenous and Health Sciences, University of Wollongong, Wollongong, NSW, Australia
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - Muinah A. Fowora
- Department of Medical Biochemistry, EKO University of Medicine and Health Sciences, Lagos, Nigeria
- Department of Molecular Biology and Biotechnology, Nigerian Institute of Medical Research, Lagos, Nigeria
| | - Cornelius A. Diya
- Department of Medical Biochemistry, EKO University of Medicine and Health Sciences, Lagos, Nigeria
| | - Faiz Elfaki
- Department of Mathematics, Physics and Statistics, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Oyelola A. Adegboye
- Public Health and Tropical Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia
| |
Collapse
|
9
|
Zhang D, Li Y, Du C, Sang L, Liu L, Li Y, Wang F, Fan W, Tang P, Zhang S, Chen D, Wang Y, Wang X, Xie X, Jiang Z, Song Y, Guo R. Evidence of pyroptosis and ferroptosis extensively involved in autoimmune diseases at the single-cell transcriptome level. J Transl Med 2022; 20:363. [PMID: 35962439 PMCID: PMC9373312 DOI: 10.1186/s12967-022-03566-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/01/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Approximately 8-9% of the world's population is affected by autoimmune diseases, and yet the mechanism of autoimmunity trigger is largely understudied. Two unique cell death modalities, ferroptosis and pyroptosis, provide a new perspective on the mechanisms leading to autoimmune diseases, and development of new treatment strategies. METHODS Using scRNA-seq datasets, the aberrant trend of ferroptosis and pyroptosis-related genes were analyzed in several representative autoimmune diseases (psoriasis, atopic dermatitis, vitiligo, multiple sclerosis, systemic sclerosis-associated interstitial lung disease, Crohn's disease, and experimental autoimmune orchitis). Cell line models were also assessed using bulk RNA-seq and qPCR. RESULTS A substantial difference was observed between normal and autoimmune disease samples involving ferroptosis and pyroptosis. In the present study, ferroptosis and pyroptosis showed an imbalance in different keratinocyte lineages of psoriatic skinin addition to a unique pyroptosis-sensitive keratinocyte subset in atopic dermatitis (AD) skin. The results also revealed that pyroptosis and ferroptosis are involved in epidermal melanocyte destruction in vitiligo. Aberrant ferroptosis has been detected in multiple sclerosis, systemic sclerosis-associated interstitial lung disease, Crohn's disease, and autoimmune orchitis. Cell line models adopted in the study also identified pro-inflammatory factors that can drive changes in ferroptosis and pyroptosis. CONCLUSION These results provide a unique perspective on the involvement of ferroptosis and pyroptosis in the pathological process of autoimmune diseases at the scRNA-seq level. IFN-γ is a critical inducer of pyroptosis sensitivity, and has been identified in two cell line models.
Collapse
Affiliation(s)
- Danfeng Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yadan Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Science, Henan Medical College of Zhengzhou University, Zhengzhou, Henan, China
- Laboratory Animal Center, School of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Chunyan Du
- Laboratory Animal Center, School of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Lina Sang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liu Liu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yingmei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fang Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenjuan Fan
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ping Tang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Sidong Zhang
- Department of Pediatric Hematology and Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dandan Chen
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yanmei Wang
- Department of Hematology, Zhengzhou People's Hospital, Zhengzhou, Henan, China
| | - Xiaoyi Wang
- Department of Pediatric Hematology and Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinsheng Xie
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Yongping Song
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Rongqun Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Academy of Medical Science, Henan Medical College of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
10
|
Zhang Y, Li Y, Cao W, Wang F, Xie X, Li Y, Wang X, Guo R, Jiang Z, Guo R. Single-Cell Analysis of Target Antigens of CAR-T Reveals a Potential Landscape of "On-Target, Off-Tumor Toxicity". Front Immunol 2022; 12:799206. [PMID: 34975912 PMCID: PMC8716389 DOI: 10.3389/fimmu.2021.799206] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/19/2021] [Indexed: 01/09/2023] Open
Abstract
Cellular immunotherapy represented by CD19-directed chimeric antigen receptor T (CAR-T) cells has achieved great success in recent years. An increasing number of CAR-T therapies are being developed for cancer treatment, but the frequent and varied adverse events, such as “on-target, off-tumor toxicity”, limit CAR-T application. Here, we identify the target antigen expression patterns of CAR therapies in 18 tissues and organs (peripheral blood mononuclear cells, bone marrow, lymph nodes, spleen, heart, ascending aortic tissue, trachea, lung, skin, kidney, bladder, esophagus, stomach, small intestine, rectum, liver, common bile duct, and pancreas) from healthy human samples. The atlas determines target antigens expressed on some normal cell types, which facilitates elucidating the cause of “on-target, off-tumor toxicity” in special tissues and organs by targeting some antigens, but not others. Moreover, we describe the target antigen expression patterns of B-lineage-derived malignant cells, acute myeloid leukemia (AML), and solid tumors. Overall, the present study indicates the pathogenesis of “on-target, off-tumor toxicity” during CAR therapies and provides guidance on taking preventive measures during CAR treatment.
Collapse
Affiliation(s)
- Yinyin Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingmei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weijie Cao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fang Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinsheng Xie
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yadan Li
- The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Xiaoyi Wang
- Department of Pediatric Hematology and Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rong Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rongqun Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Dadgar S, Mahmoudinia M, Akbari A, Zandieh E, Attaranzadeh A, Hoseinpour S, Jahanpak N, Tavanaee Sani A, Mohammadi SA. Placental infection with SARS-CoV-2, analysis of 16 cases and literature review. Arch Gynecol Obstet 2022; 305:1359-1367. [PMID: 35088195 PMCID: PMC8794612 DOI: 10.1007/s00404-021-06372-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 12/15/2021] [Indexed: 12/01/2022]
Abstract
Purpose Since December 2019, the whole world has been affected by coronavirus [severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)]. However, the effects of COVID-19 infection on pregnancy and fetal transmission are still unclear. Therefore, this study was conducted to evaluate placenta samples regarding detection of SARS-CoV-2 RNA in women affected with COVID-19.
Method This study was a part of a cohort study carried out on pregnant women with a diagnosis of COVID-19 infection who had been admitted to the Imam Reza Hospital in Mashhad, Iran, from March 20 to August 5, 2020. Clinical and laboratory information of all the patients was collected and chest computed tomography (CT) scans were reviewed. Totally, 16 placental tissue were prepared for real time polymerase chain reaction (RT-PCR) testing. All samples were tested by PowerChek PCR real-time kit (South Korea) with 2 target genes (E gene and Rd Rp gene), and Pishtaz Teb kit, (Iran) with 2 target genes (N gene and RdRp gene).
Result In the first RT-PCR kit by PowerChek kit, 6 samples were positive for a single gene (E gene) and 2 samples were positive for both genes (E gene and Rd Rp gene). In the second RT-PCR kit by Pishtaz Teb kit, 3 samples were positive for two genes (N gene and RdRp gene).
Conclusion This present study showed that infection of placenta with SARS-CoV-2 may occur in pregnancy. However, whether this infection leads to neonatal infection and serious complication in pregnancy remains unclear.
Collapse
Affiliation(s)
- Salmeh Dadgar
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihe Mahmoudinia
- Department of Obstetrics, Faculty of Medicines, Maternal and Neonatal Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Azam Akbari
- Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elaheh Zandieh
- Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Armin Attaranzadeh
- Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sajjad Hoseinpour
- Department of Molecular Genetic, Medical Laboratory, Imam Hosein Hospital, Mashhad, Iran
| | | | | | | |
Collapse
|
12
|
Knížatová N, Massányi M, Roychoudhury S, Guha P, Greifová H, Tokárová K, Jambor T, Massányi P, Lukáč N. Is there impact of the SARS-CoV-2 pandemic on steroidogenesis and fertility? Physiol Res 2021; 70:S161-S175. [PMID: 34913350 DOI: 10.33549/physiolres.934756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In December of 2019, several cases of unknown atypical respiratory diseases emerged in Wuhan, Hubei Province in China. After preliminary research, it was stated that the disease is transmittable between humans and was named COVID-19. Over the course of next months, it spread all over the world by air and sea transport and caused a global pandemic which affects life of everyone now-a-days. A large number of countries, have since been forced to take precautions such as curfews, lockdowns, wearing facemasks etc. Even with vaccines being produced in mass numbers, lack of targeted therapy continues to be a major problem. According to studies so far it seems that elderly people are more vulnerable to severe symptoms while children tend to by asymptomatic or have milder form the disease. In our review, we focused on gathering data about the virus itself, its characteristics, paths of transmission, and its effect on hormone production and secretion. In such, there is insufficient information in the literature worldwide, especially the ones that focus on the effect of COVID-19 on individual organs systems within the human body. Hence, the present evidence-based study focused on the possible effects of COVID-19 on adrenal gland and gonads i.e. on the process of steroidogenesis and fertility.
Collapse
Affiliation(s)
- N Knížatová
- Institute of Applied Biology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Nitra, Slovak Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Becker J, Qiu D, Baron W, Wilting J. Immunofluorescence studies on the expression of the SARS-CoV-2 receptors in human term placenta. Cells Tissues Organs 2021; 212:138-146. [PMID: 34915475 PMCID: PMC9148884 DOI: 10.1159/000521436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/28/2021] [Indexed: 11/29/2022] Open
Abstract
Until September 2021, the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2; COVID-19) pandemic caused over 217 million infections and over 4.5 million deaths. In pregnant women, the risk factors for the need of intensive care treatment are generally the same as in the overall population. Of note, COVID-19-positive women deliver earlier than COVID-19-negative women, and the risk for severe neonatal and perinatal morbidity and mortality is significantly higher. The probability and pathways of vertical transmission of the virus from the pregnant woman to the fetus are highly controversial. Recent data have shown that 54 (13%) of 416 neonates born to COVID-19-positive women were infected. Here, we investigated term placentas collected before the SARS-CoV-2 pandemic and studied the main COVID-19 receptors angiotensin-converting enzyme 2 (ACE2), transmembrane protease serine subtype 2 (TMPRSS2), as well as neuropilin 1 (NRP1). We performed real-time PCR and immunofluorescence on cryosections in combination with markers for syncytiotrophoblast, endothelial cells, macrophages and stromal cells. The PCR studies showed expression of both the truncated delta form of ACE2, which does not bind the COVID-19 spike protein, and the long form. The ACE2 antibody used does not distinguish between the two forms. We did not observe expression of the canonical SARS-CoV-2 entry machinery on syncytio- and cytotrophoblast. ACE2 and TMPRSS2 are co-expressed in a subpopulation of stromal cells, which in part are CD68-positive macrophages. NRP1 is localized to endothelial cells. In sum, the term placenta is not an organ that directly favors vertical transmission of COVID-19; however, microtraumas and placentitis may weaken its barrier function.
Collapse
Affiliation(s)
- Jürgen Becker
- Department of Anatomy and Cell Biology, University Medical School Göttingen, Göttingen, Germany
| | - Danny Qiu
- Department of Anatomy and Cell Biology, University Medical School Göttingen, Göttingen, Germany
| | - Walter Baron
- Department of Gynecology, Agaplesion Hospital Neu Bethlehem, Göttingen, Germany
| | - Jörg Wilting
- Department of Anatomy and Cell Biology, University Medical School Göttingen, Göttingen, Germany
- *Jörg Wilting,
| |
Collapse
|
14
|
Fuentes-Zacarías P, Murrieta-Coxca JM, Gutiérrez-Samudio RN, Schmidt A, Schmidt A, Markert UR, Morales-Prieto DM. Pregnancy and pandemics: Interaction of viral surface proteins and placenta cells. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166218. [PMID: 34311080 PMCID: PMC9188292 DOI: 10.1016/j.bbadis.2021.166218] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/06/2021] [Accepted: 07/15/2021] [Indexed: 12/18/2022]
Abstract
Throughout history, pandemics of infectious diseases caused by emerging viruses have spread worldwide. Evidence from previous outbreaks demonstrated that pregnant women are at high risk of contracting the diseases and suffering from adverse outcomes. However, while some viruses can cause major health complications for the mother and her fetus, others do not appear to affect pregnancy. Viral surface proteins bind to specific receptors on the cellular membrane of host cells and begin therewith the infection process. During pregnancy, the molecular features of these proteins may determine specific target cells in the placenta, which may explain the different outcomes. In this review, we display information on Variola, Influenza, Zika and Corona viruses focused on their surface proteins, effects on pregnancy, and possible target placental cells. This will contribute to understanding viral entry during pregnancy, as well as to develop strategies to decrease the incidence of obstetrical problems in current and future infections.
Collapse
Affiliation(s)
| | - Jose M Murrieta-Coxca
- Placenta Lab, Department of Obstetrics, Jena University Hospital, 07747 Jena, Germany
| | | | - Astrid Schmidt
- Placenta Lab, Department of Obstetrics, Jena University Hospital, 07747 Jena, Germany
| | - Andre Schmidt
- Placenta Lab, Department of Obstetrics, Jena University Hospital, 07747 Jena, Germany
| | - Udo R Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, 07747 Jena, Germany..
| | | |
Collapse
|
15
|
Mallery CS, Carrillo M, Mei A, Correia-Branco A, Kashpur O, Wallingford MC. Cellular Complexity of Hemochorial Placenta: Stem Cell Populations, Insights from scRNA-seq, and SARS-CoV-2 Susceptibility. CURRENT STEM CELL REPORTS 2021; 7:185-193. [PMID: 34697582 PMCID: PMC8527817 DOI: 10.1007/s40778-021-00194-6] [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] [Accepted: 07/15/2021] [Indexed: 11/25/2022]
Abstract
Purpose of Review The placenta is a transient organ that forms de novo and serves a critical role in supporting fetal growth and development. Placental oxygen, nutrients, and waste are transported through processes that depend on vascular structure and cell type-specific expression and localization of membrane transporters. Understanding how the placenta develops holds great significance for maternal-fetal medicine. The purpose of this review is to examine current information regarding placental progenitor populations. Recent Findings Recent advancements in single-cell RNA sequencing (scRNA-seq) provide unprecedented depth for the investigation of cell type-specific gene expression patterns in the placenta. Thus far, several mouse placenta scRNA-seq studies have been conducted which produced and analyzed transcriptomes of placental progenitors and cells of the fully developed placenta between embryonic day (E) 7.0 and E12.5. Together with human placenta scRNA-seq data which, in part, has been produced through coordinated research campaigns in the scientific community to understand the potential for SARS-CoV-2 infection, these mammalian studies lend fundamental insight into the cellular and molecular composition of hemochorial placentae found in both mouse and human. Summary Single-cell placenta research has advanced understanding of tissue-resident stem cells and molecules that are poised to support maternal-fetal communication and nutrient transport. Herein, we provide context for these recent findings by reviewing placental anatomy and cell populations, and discuss recent scRNA-seq mouse placenta findings. Further research is needed to evaluate the utility of placental stem cells in the development of new therapeutic approaches for the treatment of wound healing and disease.
Collapse
Affiliation(s)
- Christopher S. Mallery
- Tufts Medical Center, Mother Infant Research Institute, 800 Washington St, Boston, MA 02111 USA
- Texas A&M University - San Antonio, One University Way, San Antonio, 78224 USA
| | - Maira Carrillo
- Tufts Medical Center, Mother Infant Research Institute, 800 Washington St, Boston, MA 02111 USA
- Odessa College, 201 W University Blvd, Odessa, TX 79764 USA
| | - Ariel Mei
- Tufts Medical Center, Mother Infant Research Institute, 800 Washington St, Boston, MA 02111 USA
- Simmons University, 300 Fenway, Boston, MA 02115 USA
| | - Ana Correia-Branco
- Tufts Medical Center, Mother Infant Research Institute, 800 Washington St, Boston, MA 02111 USA
| | - Olga Kashpur
- Tufts Medical Center, Mother Infant Research Institute, 800 Washington St, Boston, MA 02111 USA
| | - Mary C. Wallingford
- Tufts Medical Center, Mother Infant Research Institute, 800 Washington St, Boston, MA 02111 USA
- Division of Obstetrics and Gynecology, Tufts University School of Medicine, 800 Washington Street, Boston, MA 02111 USA
| |
Collapse
|
16
|
Rad H, Röhl J, Stylianou N, Allenby M, Bazaz S, Warkiani ME, Guimaraes FSF, Clifton VL, Kulasinghe A. The Effects of COVID-19 on the Placenta During Pregnancy. Front Immunol 2021; 12:743022. [PMID: 34603330 PMCID: PMC8479199 DOI: 10.3389/fimmu.2021.743022] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 08/26/2021] [Indexed: 12/21/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a global pandemic. The virus primarily affects the lungs where it induces respiratory distress syndrome ranging from mild to acute, however, there is a growing body of evidence supporting its negative effects on other system organs that also carry the ACE2 receptor, such as the placenta. The majority of newborns delivered from SARS-CoV-2 positive mothers test negative following delivery, suggesting that there are protective mechanisms within the placenta. There appears to be a higher incidence of pregnancy-related complications in SARS-CoV-2 positive mothers, such as miscarriage, restricted fetal growth, or still-birth. In this review, we discuss the pathobiology of COVID-19 maternal infection and the potential adverse effects associated with viral infection, and the possibility of transplacental transmission.
Collapse
Affiliation(s)
- Habib Sadeghi Rad
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Joan Röhl
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Nataly Stylianou
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Mark C. Allenby
- School of Chemical Engineering, University of Queensland, St Lucia, QLD, Australia
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Sajad Razavi Bazaz
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | - Majid E. Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | | | - Vicki L. Clifton
- Mater Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - Arutha Kulasinghe
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
- The University of Queensland Diamantina Institute (UQDI), Brisbane, QLD, Australia
| |
Collapse
|
17
|
Bouachba A, Allias F, Nadaud B, Massardier J, Mekki Y, Bouscambert Duchamp M, Fourniere BDLA, Huissoud C, Trecourt A, Collardeau-Frachon S. Placental lesions and SARS-Cov-2 infection: Diffuse placenta damage associated to poor fetal outcome. Placenta 2021; 112:97-104. [PMID: 34329973 PMCID: PMC8280612 DOI: 10.1016/j.placenta.2021.07.288] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/29/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Pregnant women with covid-19 are more likely to experience preterm birth. The virus seems to be associated with a wide range of placental lesions, none of them specific. METHOD We collected cases of Covid-19 maternal infection during pregnancy associated with poor pregnancy outcomes, for which we received the placenta. We studied clinical data and described pathological findings of placenta and post-mortem examination of fetuses. We performed an immunohistochemical study and RT-PCR of SARS-Cov-2 on placenta samples. RESULTS We report 5 cases of poor fetal outcome, 3 fetal deaths and 2 extreme premature neonates, one with growth restriction, without clinical and biological sign of SARS-Cov-2 infection. All placenta presented massive perivillous fibrin deposition and large intervillous thrombi associated with strong SARS-Cov-2 expression in trophoblast and SARS-CoV-2 PCR positivity in amniotic fluid or on placenta samples. Chronic histiocytic intervillositis was present in 4/5 cases. Placental ultrasound was abnormal and the sFLT1-PIGF ratio was increased in one case. Timing between mothers' infection and the poor fetal outcome was ≤10 days in 4 cases. The massive placental damage are directly induced by the virus whose receptors are expressed on trophoblast, leading to trophoblast necrosis and massive inflammation in villous chamber, in a similar way it occurs in diffuse alveolar damage in adults infected by SARS-Cov-2. DISCUSSION SARS-Cov-2 can be associated to a rare set of placental lesions which can lead to fetal demise, preterm birth, or growth restriction. Stronger surveillance of mothers infected by SARS-Cov-2 is required.
Collapse
Affiliation(s)
- Amine Bouachba
- Institut de Pathologie Multisite des Hospices Civils de Lyon, 69677, Bron Cedex, Lyon, France; SOFFOET-Société Française de Fœtopathologie, 75014, Paris, France.
| | - Fabienne Allias
- SOFFOET-Société Française de Fœtopathologie, 75014, Paris, France; Institut de Pathologie, Centre hospitalier Lyon Sud, Hospices Civils de Lyon, 69495, Pierre Bénite, Lyon, France
| | - Beatrice Nadaud
- Institut de Pathologie Multisite des Hospices Civils de Lyon, 69677, Bron Cedex, Lyon, France; SOFFOET-Société Française de Fœtopathologie, 75014, Paris, France
| | - Jerome Massardier
- SOFFOET-Société Française de Fœtopathologie, 75014, Paris, France; Service de gynécologie-obstétrique de l'Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, 69677, Bron Cedex, Lyon, France
| | - Yahia Mekki
- Institut des Agents Infectieux des Hospices Civils de Lyon, Service de Virologie, 69004, Lyon, France
| | - Maude Bouscambert Duchamp
- Institut des Agents Infectieux des Hospices Civils de Lyon, Service de Virologie, 69004, Lyon, France
| | - Benoit De LA Fourniere
- Service de Gynécologie-obstétrique de La Croix-Rousse, Hospices Civils de Lyon, 69004, Lyon, France
| | - Cyril Huissoud
- Service de Gynécologie-obstétrique de La Croix-Rousse, Hospices Civils de Lyon, 69004, Lyon, France; Université Claude Bernard Lyon 1, Faculté de Médecine Lyon Est, 69008, France
| | - Alexis Trecourt
- Institut de Pathologie Multisite des Hospices Civils de Lyon, 69677, Bron Cedex, Lyon, France
| | - Sophie Collardeau-Frachon
- Institut de Pathologie Multisite des Hospices Civils de Lyon, 69677, Bron Cedex, Lyon, France; SOFFOET-Société Française de Fœtopathologie, 75014, Paris, France; Université Claude Bernard Lyon 1, Faculté de Médecine Lyon Est, 69008, France
| |
Collapse
|
18
|
COVID-19 in Pregnancy-Perinatal Outcomes and Vertical Transmission Preventative Strategies, When Considering More Transmissible SARS-CoV-2 Variants. J Clin Med 2021; 10:jcm10163724. [PMID: 34442020 PMCID: PMC8397094 DOI: 10.3390/jcm10163724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/05/2021] [Accepted: 08/19/2021] [Indexed: 11/16/2022] Open
Abstract
The COVID-19 pandemic affected the physical and mental health of people around the world and left unprepared health care systems struggling to mount an adequate response. Understanding the impact of COVID-19 on pregnancy in terms of perinatal and fetal outcomes is essential to propose strategies for mminimising viral transmission. Overall, 91 pregnant women in labour, or with indication for induction of labour, with COVID-19 were admitted to hospital. On the day of admission, each pregnant woman underwent a nasopharyngeal swab to validate SARS-CoV-2 infection. Whenever delivery was by caesarean section, an amniotic fluid sample was collected after uterus incision. Neonates were tested twice: first by nasopharyngeal swab at birth and secondly either at 24 h after (when babies were isolated) or at discharge (when rooming-in). All samples underwent rRT-PCR testing for SARS-CoV-2. The SARS-CoV-2 RNA tests by nasopharyngeal swab of the pregnant women produced positive results in 47 patients. This cohort gave birth to 48 infants who were double tested by nasopharyngeal swab and included in the prospective observational study. Moreover, in this same cohort, 39 amniotic fluid samples were taken during caesarean section. All samples underwent rRT-PCR testing for SARS-CoV-2 and came back negative. The study results suggest a low risk of vertical transmission of COVID-19 and favourable perinatal outcomes due to adequate preventative strategies. This approach may prove to be more beneficial in the new SARS-CoV-2 variants era.
Collapse
|
19
|
Anifandis G, Messini CI, Simopoulou M, Sveronis G, Garas A, Daponte A, Messinis IE. SARS-CoV-2 vs. human gametes, embryos and cryopreservation. Syst Biol Reprod Med 2021; 67:260-269. [PMID: 34060390 DOI: 10.1080/19396368.2021.1922537] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The COVID-19 pandemic, caused by the SARS-CoV-2 virus, is an unprecedented global situation, and all countries have adopted their own measurements to mitigate the spread of the virus in the first as well as in the subsequent waves of infection. All measures, especially in the first wave of the pandemic, were in combination with recommendations provided by professional and scientific organizations. Similar measures were applied to specific procedures, such as the management of infertility, including in vitro fertilization-embryo transfer (IVF-ET) treatments. Although there is no clear scientific evidence yet that the SARS-CoV-2 may exert negative effects on IVF outcome, especially at the early stages, several clinical reports indicate that the virus may impact male fertility through specific receptors presented at the somatic cells of the testis and used by the virus in order to gain entry to the respective cells. Nevertheless, it is not unreasonable to suspect that the virus may affect sperm function as well as oocyte performance directly through specific receptors or indirectly through other signaling pathways. Despite the good practice of IVF laboratory techniques, culture media may also be contaminated during equilibration when airborne virus's particles can contaminate culture media from an already infected embryology area or staff. Furthermore, although there is no clinical evidence, liquid nitrogen could be a route of infection for gametes and embryos when it has been contaminated during production or transportation. Therefore, cryopreservation of gametes and embryos must be virus-free. This communication aims to provide some aspects of the possible impact of the virus on gametes and embryos and how it may affect the cryopreservation procedures.Abbreviations: ACE2: angiotensin- converting enzyme 2; ART: assisted reproductive technology; ASRM: American Society for Reproductive Medicine; CDC: Centers for Disease Control and Prevention; COVID-19: coronavirus disease 2019; ESHRE: European Society of Human Reproduction and Embryology; ET: embryo transfer; FSH: follicle stimulating hormone; IFFS: International Federation of Fertility Societies; IVF: in vitro fertilization; LH: luteinizing hormone; LN: liquid nitrogen; SARS-CoV-2: severe acute respiratory syndrome coronavirus 2; T: testosterone; WHO: World Health Organization.
Collapse
Affiliation(s)
- George Anifandis
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Christina I Messini
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Mara Simopoulou
- Department of Physiology, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - George Sveronis
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece.,Department of Obstetrics and Gynaecology, Norfolk and Norwich University Hospitals, Norwich Medical School, University of East Anglia, Norwich, UK
| | - Antonios Garas
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Alexandros Daponte
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Ioannis E Messinis
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| |
Collapse
|
20
|
Cimolai N. A Comprehensive Analysis of Maternal and Newborn Disease and Related Control for COVID-19. SN COMPREHENSIVE CLINICAL MEDICINE 2021; 3:1272-1294. [PMID: 33754135 PMCID: PMC7968576 DOI: 10.1007/s42399-021-00836-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Accepted: 02/22/2021] [Indexed: 02/07/2023]
Abstract
The maternal-fetal/newborn unit is established at risk for COVID-19 infection. This narrative review summarizes the contemporary and cumulative publications which detail maternal infection, antenatal and newborn infections, and maternal/fetal/newborn management and prevention. There is a wide spectrum of maternal disease, but the potential for severe disease albeit in a minority is confirmed. COVID-19 carries risk for preterm delivery. Pregnant females can suffer multisystem disease, and co-morbidities play a significant role in risk. Congenital infection has been supported by several anecdotal reports, but strong confirmatory data are few. No typical congenital dysmorphisms are evident. Nevertheless, placental vascular compromise must be considered a risk for the fetus during advanced maternal infections. Clinical manifestations of newborn infection have been mild to moderate and relatively uncommon. Proven antiviral therapy is of yet lacking. The mode of delivery is a medical decision that must include patient risk assessment and patient directives. Both presymptomatic and asymptomatic mothers and offspring can complicate infection control management with the potential for spread to others in several regards. In the interim, infections of the maternal-fetal-newborn unit must be taken seriously both for the disease so caused and the potential for further dissemination of disease.
Collapse
Affiliation(s)
- Nevio Cimolai
- Faculty of Medicine, The University of British Columbia, Vancouver, Canada
- Children’s and Women’s Health Centre of British Columbia, 4480 Oak Street, Vancouver, B.C. V6H3V4 Canada
| |
Collapse
|
21
|
Guo R, Lü M, Cao F, Wu G, Gao F, Pang H, Li Y, Zhang Y, Xing H, Liang C, Lyu T, Du C, Li Y, Guo R, Xie X, Li W, Liu D, Song Y, Jiang Z. Single-cell map of diverse immune phenotypes in the acute myeloid leukemia microenvironment. Biomark Res 2021; 9:15. [PMID: 33648605 PMCID: PMC7919996 DOI: 10.1186/s40364-021-00265-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/04/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Knowledge of immune cell phenotypes, function, and developmental trajectory in acute myeloid leukemia (AML) microenvironment is essential for understanding mechanisms of evading immune surveillance and immunotherapy response of targeting special microenvironment components. METHODS Using a single-cell RNA sequencing (scRNA-seq) dataset, we analyzed the immune cell phenotypes, function, and developmental trajectory of bone marrow (BM) samples from 16 AML patients and 4 healthy donors, but not AML blasts. RESULTS We observed a significant difference between normal and AML BM immune cells. Here, we defined the diversity of dendritic cells (DC) and macrophages in different AML patients. We also identified several unique immune cell types including T helper cell 17 (TH17)-like intermediate population, cytotoxic CD4+ T subset, T cell: erythrocyte complexes, activated regulatory T cells (Treg), and CD8+ memory-like subset. Emerging AML cells remodels the BM immune microenvironment powerfully, leads to immunosuppression by accumulating exhausted/dysfunctional immune effectors, expending immune-activated types, and promoting the formation of suppressive subsets. CONCLUSION Our results provide a comprehensive AML BM immune cell census, which can help to select pinpoint targeted drug and predict efficacy of immunotherapy.
Collapse
Affiliation(s)
- Rongqun Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengdie Lü
- Joint National Laboratory for Antibody Drug Engineering, Key Laboratory of Cellular and Molecular Immunology of Henan Province, Institute of Translational Medicine, School of Basic Medicine, Henan University, Kaifeng, Henan, China
| | - Fujiao Cao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Guanghua Wu
- The Academy of Medical Science, College of Medical, Zhengzhou University, Zhengzhou, Henan, China
| | - Fengcai Gao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haili Pang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yadan Li
- The Academy of Medical Science, College of Medical, Zhengzhou University, Zhengzhou, Henan, China
- The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Yinyin Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haizhou Xing
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chunyan Liang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tianxin Lyu
- The Academy of Medical Science, College of Medical, Zhengzhou University, Zhengzhou, Henan, China
- The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Chunyan Du
- Laboratory Animal Center, School of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yingmei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Rong Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xinsheng Xie
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Delong Liu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Yongping Song
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
22
|
Sanchez J, Vigil-De Gracia P, Guerrero E, Gaitán M, Fu C, Chen-Germán M, Villalobos R, Coronado L, Martínez AA, Araúz D, Saenz L, Chavarría O, Góndola J, Moreno A, González C, Vega S, Campana S, Ng Chinkee J, López-Vergès S, Solís MA. Severe acute respiratory syndrome coronavirus 2 detected in placentas of 2 coronavirus disease 2019-positive asymptomatic pregnant women-case report. AJOG GLOBAL REPORTS 2021; 1:100001. [PMID: 33842905 PMCID: PMC7838577 DOI: 10.1016/j.xagr.2020.100001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
There is limited evidence regarding severe acute respiratory syndrome coronavirus 2 infection in the placenta of pregnant women who tested positive, and if this could be a route for vertical transmission of the virus in utero. We present the cases of 2 pregnant women in their third trimester who were admitted for delivery by cesarean delivery and who, through universal screening, tested positive for coronavirus disease 2019. The maternal and fetal sides of the placenta were sectioned from both patients for viral analysis. Real-time polymerase chain reaction analysis of the placental-extracted RNA revealed a severe acute respiratory syndrome coronavirus 2 infection on the fetal side of the placenta in both patients. The virus was isolated from the patient with the lowest cycle threshold value on the fetal side of the placenta. Whole genome sequencing showed that the virus detected in this placenta was from the B1 lineage. Immunohistochemical analysis of the placental tissue detected severe acute respiratory syndrome coronavirus 2 in the endothelial cells of chorionic villi vessels proximal to both the maternal and fetal sides, with a granular cytoplasmic pattern and perinuclear reinforcement. Histologic examination of the placenta also detected a dense infiltrate of lymphoid cells around decidual vessels and endothelial cells with cytopathic changes, especially on the maternal side. Nasopharyngeal swabs from the infants that were subjected to reverse transcription quantitative polymerase chain reaction testing were negative for severe acute respiratory syndrome coronavirus 2 at 24 hours after birth. A follow-up analysis of the infants for immunoglobin G and immunoglobin M expression, clinical manifestations, and long-term developmental abnormalities is recommended.
Collapse
Affiliation(s)
- Jaime Sanchez
- Department of Gynecology and Obstetrics, Hospital Santo Tomás, Panama City, Republic of Panama (Dr Sanchez)
| | - Paulino Vigil-De Gracia
- Division of Gynecology and Obstetrics, Complejo Hospitalario Metropolitano Dr Arnulfo Arias Madrid, Panama City, Republic of Panama (Dr Vigil-De Gracia, Dr Campana, and Dr Chinkee).,Sistema Nacional de Investigación, SENACYT, Panama City, Republic of Panama (Dr Vigil-De Gracia, Dr Martínez and Drs López-Vergès and Solís)
| | - Erika Guerrero
- Stem Cell Research Group, Department of Sexual and Reproductive Health Research, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama (Dr Guerrero, Ms Fu, and Ms Vega and Dr Solis)
| | - Melissa Gaitán
- Department of Virology and Biotechnology Research, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama (Ms Gaitán, Ms Chen-Germán, Ms Araúz, Saenz and Dr López-Vergès)
| | - Cindy Fu
- Stem Cell Research Group, Department of Sexual and Reproductive Health Research, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama (Dr Guerrero, Ms Fu, and Ms Vega and Dr Solis)
| | - María Chen-Germán
- Department of Virology and Biotechnology Research, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama (Ms Gaitán, Ms Chen-Germán, Ms Araúz, Saenz and Dr López-Vergès)
| | - Rodrigo Villalobos
- Department of Diagnostics, Pathology Service, Hospital Santo Tomás, Panama City, Republic of Panama (Dr Villalobos and Dr Coronado)
| | - Luis Coronado
- Department of Diagnostics, Pathology Service, Hospital Santo Tomás, Panama City, Republic of Panama (Dr Villalobos and Dr Coronado)
| | - Alexander A Martínez
- Department of Genomics and Proteomics Research, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama (Dr Martínez, Ms Chavarría, Ms Góndola, Ms Moreno and Ms González).,Universidad de Panamá, Panama City, Republic of Panama (Dr Martínez, Ms González and Dr López-Vergès and Dr Solis).,Sistema Nacional de Investigación, SENACYT, Panama City, Republic of Panama (Dr Vigil-De Gracia, Dr Martínez and Drs López-Vergès and Solís)
| | - Dimelza Araúz
- Department of Virology and Biotechnology Research, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama (Ms Gaitán, Ms Chen-Germán, Ms Araúz, Saenz and Dr López-Vergès)
| | - Lisseth Saenz
- Department of Virology and Biotechnology Research, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama (Ms Gaitán, Ms Chen-Germán, Ms Araúz, Saenz and Dr López-Vergès)
| | - Oris Chavarría
- Department of Genomics and Proteomics Research, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama (Dr Martínez, Ms Chavarría, Ms Góndola, Ms Moreno and Ms González)
| | - Jessica Góndola
- Department of Genomics and Proteomics Research, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama (Dr Martínez, Ms Chavarría, Ms Góndola, Ms Moreno and Ms González)
| | - Ambar Moreno
- Department of Genomics and Proteomics Research, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama (Dr Martínez, Ms Chavarría, Ms Góndola, Ms Moreno and Ms González)
| | - Claudia González
- Department of Genomics and Proteomics Research, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama (Dr Martínez, Ms Chavarría, Ms Góndola, Ms Moreno and Ms González).,Universidad de Panamá, Panama City, Republic of Panama (Dr Martínez, Ms González and Dr López-Vergès and Dr Solis)
| | - Shantal Vega
- Stem Cell Research Group, Department of Sexual and Reproductive Health Research, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama (Dr Guerrero, Ms Fu, and Ms Vega and Dr Solis)
| | - Sara Campana
- Division of Gynecology and Obstetrics, Complejo Hospitalario Metropolitano Dr Arnulfo Arias Madrid, Panama City, Republic of Panama (Dr Vigil-De Gracia, Dr Campana, and Dr Chinkee)
| | - Jorge Ng Chinkee
- Division of Gynecology and Obstetrics, Complejo Hospitalario Metropolitano Dr Arnulfo Arias Madrid, Panama City, Republic of Panama (Dr Vigil-De Gracia, Dr Campana, and Dr Chinkee)
| | - Sandra López-Vergès
- Department of Virology and Biotechnology Research, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama (Ms Gaitán, Ms Chen-Germán, Ms Araúz, Saenz and Dr López-Vergès).,Universidad de Panamá, Panama City, Republic of Panama (Dr Martínez, Ms González and Dr López-Vergès and Dr Solis).,Sistema Nacional de Investigación, SENACYT, Panama City, Republic of Panama (Dr Vigil-De Gracia, Dr Martínez and Drs López-Vergès and Solís)
| | - Mairim Alexandra Solís
- Department of Virology and Biotechnology Research, Gorgas Memorial Institute for Health Studies, Panama City, Republic of Panama (Ms Gaitán, Ms Chen-Germán, Ms Araúz, Saenz and Dr López-Vergès).,Sistema Nacional de Investigación, SENACYT, Panama City, Republic of Panama (Dr Vigil-De Gracia, Dr Martínez and Drs López-Vergès and Solís)
| |
Collapse
|
23
|
Thomas N, Gurvich C, Kulkarni J. Sex Differences and COVID-19. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1327:79-91. [PMID: 34279830 DOI: 10.1007/978-3-030-71697-4_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Biological sex and psychosocial gender both play a role in many disease outcomes, and the novel coronavirus disease (COVID-19) is no different. Clinical observations in COVID-19 patient data delineate clear disparities between males and females, indicating males are at a higher risk for poorer disease outcomes. Although we are yet to understand the sex and gender-based disparities specific to COVID-19, there is evidence for sex-based differences in the endocrine, immune and renin-angiotensin system, all systems implicated in COVID-19 outcomes. Such disparities are largely thought to be driven by sex chromosomes and modulating sex hormones, which are known to vary between sex, and across the reproductive lifespan. Understanding and exploiting these driving factors are critical to understanding the pathobiology of SARS-CoV-2 virus and may lead to the development of novel therapies and increase the efficacy of preventative vaccine strategies currently under development. This chapter focuses on the endocrine, immune and renin-angiotensin system and genetic sex-based differences that could account for the meaningful differences observed in the outcomes of the SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Natalie Thomas
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Australia. .,Central Clinical School, Monash University, Clayton, VIC, Australia.
| | - Caroline Gurvich
- Central Clinical School, Monash University, Clayton, VIC, Australia
| | | |
Collapse
|