1
|
Wang L, Mu Q, Zhang W, Zheng W, Zhu X, Yu Y, Wang Y, Xu W, Lu Z, Han X. Placental targeted drug delivery: a review of recent progress. NANOSCALE 2025; 17:8316-8335. [PMID: 40070242 DOI: 10.1039/d4nr05338a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
The placenta plays a crucial role in mediating nutrient and gas exchange between the mother and fetus during pregnancy. Targeting therapeutic agents to the placenta presents significant opportunities for treating placental disorders and enhancing fetal outcomes. However, the unique structural complexity and selective permeability of the placenta pose substantial challenges for effective drug delivery. This review provides a comprehensive overview of current strategies for placental targeting, including lipid nanoparticle (LNP) delivery systems, targeted peptide modifications, specific antibody targeting of placental receptors, and the use of viral vectors. We critically analyze the advantages and limitations of each approach, emphasizing recent advancements in enhancing targeting specificity and delivery efficiency. By consolidating the latest research developments, this review aims to foster further innovation in placental drug delivery methods and contribute significantly to the advancement of therapeutic strategies for placental disorders, ultimately improving outcomes for both mother and fetus.
Collapse
Affiliation(s)
- Linjian Wang
- Department of Obstetrics, Haining Maternal and Child Health Hospital, Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, No. 309, East Shuiyueting Road, Xiashi Street, Haining, Zhejiang, 314400, China
| | - Qiuqiu Mu
- Third Affliated Hospital of Wenzhou Medical University, WanSong Road No. 108, Ruian, Wenzhou, Zhejiang, 325200, China
| | - Wenjing Zhang
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| | - Weiqian Zheng
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| | - Xiaojun Zhu
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| | - Ying Yu
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| | - YuPeng Wang
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| | - Wenli Xu
- Department of Obstetrics, Haining Maternal and Child Health Hospital, Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, No. 309, East Shuiyueting Road, Xiashi Street, Haining, Zhejiang, 314400, China
| | - Zhimin Lu
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| | - Xiujun Han
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Xueshi Road No. 1, Hangzhou, Zhejiang, 310006, China.
| |
Collapse
|
2
|
Pesch MH, Mowers J, Huynh A, Schleiss MR. Intrauterine Fetal Demise, Spontaneous Abortion and Congenital Cytomegalovirus: A Systematic Review of the Incidence and Histopathologic Features. Viruses 2024; 16:1552. [PMID: 39459885 PMCID: PMC11512218 DOI: 10.3390/v16101552] [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: 06/30/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
The objective was to review the existing literature reporting on spontaneous abortion (SA) and intrauterine fetal demise (IUFD) associated with cytomegalovirus (CMV) infection. A review using standardized terminology such as 'intrauterine fetal death', 'congenital cytomegalovirus' and 'CMV' was performed using PubMed and Embase (Medline) using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) methodology. Twenty-one studies met inclusion criteria. CMV was identified as a potential or likely factor in a median of 7.1% of SA or IUFD in study cohorts. Of the studies, 11 used fetal remains, 18 used placenta, 6 used serum, and 1 used post-mortem dried blood spot as specimens for testing for CMV. Features commonly observed were fetal thrombotic vasculopathy, hydrops fetalis and chronic villitis. CMV is frequently noted in studies evaluating viral etiologies of SA or IUFD. Large population-based studies are needed to estimate the incidence of CMV-associated SA or IUFD. CMV and congenital CMV should be included on the differential diagnosis in all cases of SA or IUFD of unknown etiology.
Collapse
Affiliation(s)
- Megan H. Pesch
- Division of Developmental and Behavioral Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jonathan Mowers
- Division of Pathology, Ascension Hospital Providence, Southfield, MI 48075, USA;
| | - Anh Huynh
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA;
| | - Mark R. Schleiss
- Division of Pediatric Infectious Diseases, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
3
|
Sanami S, Shamsabadi S, Dayhimi A, Pirhayati M, Ahmad S, Pirhayati A, Ajami M, Hemati S, Shirvani M, Alagha A, Abbarin D, Alizadeh A, Pazoki-Toroudi H. Association between cytomegalovirus infection and neurological disorders: A systematic review. Rev Med Virol 2024; 34:e2532. [PMID: 38549138 DOI: 10.1002/rmv.2532] [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: 01/30/2024] [Revised: 03/11/2024] [Accepted: 03/14/2024] [Indexed: 04/02/2024]
Abstract
Cytomegalovirus (CMV) belongs to the Herpesviridae family and is also known as human herpesvirus type 5. It is a common virus that usually doesn't cause any symptoms in healthy individuals. However, once infected, the virus remains in the host's body for life and can reactivate when the host's immune system weakens. This virus has been linked to several neurological disorders, including Alzheimer's disease, Parkinson's disease, Autism spectrum disorder, Huntington's disease (HD), ataxia, Bell's palsy (BP), and brain tumours, which can cause a wide range of symptoms and challenges for those affected. CMV may influence inflammation, contribute to brain tissue damage, and elevate the risk of moderate-to-severe dementia. Multiple studies suggest a potential association between CMV and ataxia in various conditions, including Guillain-Barré syndrome, chronic inflammatory demyelinating polyneuropathy, acute cerebellitis, etc. On the other hand, the evidence regarding CMV involvement in BP is conflicting, and also early indications of a link between CMV and HD were challenged by subsequent research disproving CMV's presence. This systematic review aims to comprehensively investigate any link between the pathogenesis of CMV and its potential role in neurological disorders and follows the preferred reporting items for systematic review and meta-analysis checklist. Despite significant research into the potential links between CMV infection and various neurological disorders, the direct cause-effect relationship is not fully understood and several gaps in knowledge persist. Therefore, continued research is necessary to gain a better understanding of the role of CMV in neurological disorders and potential treatment avenues.
Collapse
Affiliation(s)
- Samira Sanami
- Abnormal Uterine Bleeding Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Shahnam Shamsabadi
- Department of Physiology, Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Dayhimi
- Department of Physiology, Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Pirhayati
- Psychiatric Department, Rasool Akram Hospital, Iran University of Medical Science, Tehran, Iran
| | - Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Pakistan
- Department of Computer Sciences, Virginia Tech, Blacksburg, Virginia, USA
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| | | | - Marjan Ajami
- National Nutrition and Food Technology Research Institute, School of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Hemati
- Department of Environmental Health Engineering, School of Health, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Masoud Shirvani
- Department of Neurosurgery, Salamat-Farda Hospital, Tehran, Iran
| | - Ahmad Alagha
- Department of Neurosurgery, Salamat-Farda Hospital, Tehran, Iran
| | - Davood Abbarin
- Department of Neurosurgery, Salamat-Farda Hospital, Tehran, Iran
| | - Akram Alizadeh
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Hamidreza Pazoki-Toroudi
- Department of Physiology, Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Carvalho TP, Toledo FAO, Bautista DFA, Silva MF, Oliveira JBS, Lima PA, Costa FB, Ribeiro NQ, Lee JY, Birbrair A, Paixão TA, Tsolis RM, Santos RL. Pericytes modulate endothelial inflammatory response during bacterial infection. mBio 2024; 15:e0325223. [PMID: 38289074 PMCID: PMC10936204 DOI: 10.1128/mbio.03252-23] [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: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 03/14/2024] Open
Abstract
Pericytes are located around blood vessels, in close contact with endothelial cells. We discovered that pericytes dampen pro-inflammatory endothelial cell responses. Endothelial cells co-cultured with pericytes had markedly reduced expression of adhesion molecules (PECAM-1 and ICAM-1) and proinflammatory cytokines (CCL-2 and IL-6) in response to bacterial stimuli (Brucella ovis, Listeria monocytogenes, or Escherichia coli lipopolysaccharide). Pericyte-depleted mice intraperitoneally inoculated with either B. ovis, a stealthy pathogen that does not trigger detectable inflammation, or Listeria monocytogenes, developed peritonitis. Further, during Citrobacter rodentium infection, pericyte-depleted mice developed severe intestinal inflammation, which was not evident in control mice. The anti-inflammatory effect of pericytes required connexin 43, as either chemical inhibition or silencing of connexin 43 abrogated pericyte-mediated suppression of endothelial inflammatory responses. Our results define a mechanism by which pericytes modulate inflammation during infection, which shifts our understanding of pericyte biology: from a structural cell to a pro-active player in modulating inflammation. IMPORTANCE A previously unknown mechanism by which pericytes modulate inflammation was discovered. The absence of pericytes or blocking interaction between pericytes and endothelium through connexin 43 results in stronger inflammation, which shifts our understanding of pericyte biology, from a structural cell to a player in controlling inflammation.
Collapse
Affiliation(s)
- Thaynara P. Carvalho
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| | - Frank A. O. Toledo
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Diego F. A. Bautista
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Monique F. Silva
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jefferson B. S. Oliveira
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Pâmela A. Lima
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fabíola B. Costa
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Noelly Q. Ribeiro
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jee-Yon Lee
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| | - Alexander Birbrair
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Tatiane A. Paixão
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Reneé M. Tsolis
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| | - Renato L. Santos
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| |
Collapse
|
5
|
Lawrence SM, Goshia T, Sinha M, Fraley SI, Williams M. Decoding human cytomegalovirus for the development of innovative diagnostics to detect congenital infection. Pediatr Res 2024; 95:532-542. [PMID: 38146009 PMCID: PMC10837078 DOI: 10.1038/s41390-023-02957-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 11/14/2023] [Accepted: 11/27/2023] [Indexed: 12/27/2023]
Abstract
Cytomegalovirus is the most common cause of congenital infectious disease and the leading nongenetic etiology of sensorineural hearing loss. Although most infected neonates are asymptomatic at birth, congenital cytomegalovirus infection is responsible for nearly 400 infant deaths annually in the United States and may lead to significant long-term neurodevelopmental impairments in survivors. The resulting financial and social burdens of congenital cytomegalovirus infection have led many medical centers to initiate targeted testing after birth, with a growing advocacy to advance universal newborn screening. While no cures or vaccines are currently available to eliminate or prevent cytomegalovirus infection, much has been learned over the last five years regarding disease pathophysiology and viral replication cycles that may enable the development of innovative diagnostics and therapeutics. This Review will detail our current understanding of congenital cytomegalovirus infection, while focusing our discussion on routine and emerging diagnostics for viral detection, quantification, and long-term prognostication. IMPACT: This review highlights our current understanding of the fetal transmission of human cytomegalovirus. It details clinical signs and physical findings of congenital cytomegalovirus infection. This submission discusses currently available cytomegalovirus diagnostics and introduces emerging platforms that promise improved sensitivity, specificity, limit of detection, viral quantification, detection of genomic antiviral resistance, and infection staging (primary, latency, reactivation, reinfection).
Collapse
Affiliation(s)
- Shelley M Lawrence
- University of Utah, College of Medicine, Department of Pediatrics, Division of Neonatology, Salt Lake City, UT, USA.
| | - Tyler Goshia
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | | | - Stephanie I Fraley
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Marvin Williams
- University of Oklahoma, College of Medicine, Department of Obstetrics and Gynecology, Division of Fetal-Maternal Medicine, Oklahoma City, OK, USA
| |
Collapse
|
6
|
Al-Beltagi M, Saeed NK, Elbeltagi R, Bediwy AS, Aftab SAS, Alhawamdeh R. Viruses and autism: A Bi-mutual cause and effect. World J Virol 2023; 12:172-192. [PMID: 37396705 PMCID: PMC10311578 DOI: 10.5501/wjv.v12.i3.172] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/16/2023] [Accepted: 04/18/2023] [Indexed: 06/21/2023] Open
Abstract
Autism spectrum disorder (ASD) is a group of heterogeneous, multi-factorial, neurodevelopmental disorders resulting from genetic and environmental factors interplay. Infection is a significant trigger of autism, especially during the critical developmental period. There is a strong interplay between the viral infection as a trigger and a result of ASD. We aim to highlight the mutual relationship between autism and viruses. We performed a thorough literature review and included 158 research in this review. Most of the literature agreed on the possible effects of the viral infection during the critical period of development on the risk of developing autism, especially for specific viral infections such as Rubella, Cytomegalovirus, Herpes Simplex virus, Varicella Zoster Virus, Influenza virus, Zika virus, and severe acute respiratory syndrome coronavirus 2. Viral infection directly infects the brain, triggers immune activation, induces epigenetic changes, and raises the risks of having a child with autism. At the same time, there is some evidence of increased risk of infection, including viral infections in children with autism, due to lots of factors. There is an increased risk of developing autism with a specific viral infection during the early developmental period and an increased risk of viral infections in children with autism. In addition, children with autism are at increased risk of infection, including viruses. Every effort should be made to prevent maternal and early-life infections and reduce the risk of autism. Immune modulation of children with autism should be considered to reduce the risk of infection.
Collapse
Affiliation(s)
- Mohammed Al-Beltagi
- Department of Pediatrics, Faculty of Medicine, Tanta University, Tanta 31511, Alghrabia, Egypt
- Department of Pediatrics, University Medical Center, King Abdulla Medical City, Dr. Sulaiman Al Habib Medical Group, Manama 26671, Bahrain
| | - Nermin Kamal Saeed
- Medical Microbiology Section, Pathology Department, Salmaniya Medical Complex, Ministry of Health, Kingdom of Bahrain, Manama 12, Bahrain
- Microbiology Section, Pathology Department, Irish Royal College of Surgeon, Busaiteen 15503, Muharraq, Bahrain
| | - Reem Elbeltagi
- Department of Medicine, The Royal College of Surgeons in Ireland - Bahrain, Busiateen 15503, Muharraq, Bahrain
| | - Adel Salah Bediwy
- Department of Pulmonolgy, Faculty of Medicine, Tanta University, Tanta 31527, Alghrabia, Egypt
- Department of Chest Disease, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Dr. Sulaiman Al Habib Medical Group, Manama 26671, Bahrain
| | - Syed A Saboor Aftab
- Endocrinology and DM, William Harvey Hospital (Paula Carr Centre), Ashford TN24 0LZ, Kent, United Kingdom
| | - Rawan Alhawamdeh
- Pediatrics Research and Development, Genomics Creativity and Play Center, Manama 0000, Bahrain
| |
Collapse
|
7
|
Kumar M, Saadaoui M, Al Khodor S. Infections and Pregnancy: Effects on Maternal and Child Health. Front Cell Infect Microbiol 2022; 12:873253. [PMID: 35755838 PMCID: PMC9217740 DOI: 10.3389/fcimb.2022.873253] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/04/2022] [Indexed: 12/22/2022] Open
Abstract
Pregnancy causes physiological and immunological adaptations that allow the mother and fetus to communicate with precision in order to promote a healthy pregnancy. At the same time, these adaptations may make pregnant women more susceptible to infections, resulting in a variety of pregnancy complications; those pathogens may also be vertically transmitted to the fetus, resulting in adverse pregnancy outcomes. Even though the placenta has developed a robust microbial defense to restrict vertical microbial transmission, certain microbial pathogens have evolved mechanisms to avoid the placental barrier and cause congenital diseases. Recent mechanistic studies have begun to uncover the striking role of the maternal microbiota in pregnancy outcomes. In this review, we discuss how microbial pathogens overcome the placental barrier to cause congenital diseases. A better understanding of the placental control of fetal infection should provide new insights into future translational research.
Collapse
Affiliation(s)
- Manoj Kumar
- Research Department, Sidra Medicine, Doha, Qatar
| | | | | |
Collapse
|
8
|
Girsch JH, Mejia Plazas MC, Olivier A, Farah M, Littlefield D, Behl S, Punia S, Sakemura R, Hemsath JR, Norgan A, Enninga EAL, Johnson EL, Chakraborty R. Host-Viral Interactions at the Maternal-Fetal Interface. What We Know and What We Need to Know. FRONTIERS-A JOURNAL OF WOMEN STUDIES 2022; 2:833106. [PMID: 36742289 PMCID: PMC9894500 DOI: 10.3389/fviro.2022.833106] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In humans, the hemochorial placenta is a unique temporary organ that forms during pregnancy to support fetal development, gaseous exchange, delivery of nutrition, removal of waste products, and provides immune protection, while maintaining tolerance to the HLA-haploidentical fetus. In this review, we characterize decidual and placental immunity during maternal viral (co)-infection with HIV-1, human cytomegalovirus (HCMV), and Zika virus. We discuss placental immunology, clinical presentation, and epidemiology, before characterizing host susceptibility and cellular tropism, and how the three viruses gain access into specific placental target cells. We describe current knowledge on host-viral interactions with decidual and stromal human placental macrophages or Hofbauer cells, trophoblasts including extra villous trophoblasts, T cells, and decidual natural killer (dNK) cells. These clinically significant viral infections elicit both innate and adaptive immune responses to control replication. However, the three viruses either during mono- or co-infection (HIV-1 and HCMV) escape detection to initiate placental inflammation associated with viral transmission to the developing fetus. Aside from congenital or perinatal infection, other adverse pregnancy outcomes include preterm labor and spontaneous abortion. In addition, maternal HIV-1 and HCMV co-infection are associated with impaired fetal and infant immunity in postnatal life and poor clinical outcomes during childhood in exposed infants, even in the absence of vertical transmission of HIV-1. Given the rapidly expanding numbers of HIV-1-exposed uninfected infants and children globally, further research is urgently needed on neonatal immune programming during maternal mono-and co-infection. This review therefore includes sections on current knowledge gaps that may prompt future research directions. These gaps reflect an emerging but poorly characterized field. Their significance and potential investigation is underscored by the fact that although viral infections result in adverse consequences in both mother and developing fetus/newborn, antiviral and immunomodulatory therapies can improve clinical outcomes in the dyad.
Collapse
Affiliation(s)
- James H. Girsch
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States,,Mayo Clinic Graduate School of Biomedical Science, Rochester, MN, United States
| | - Maria C. Mejia Plazas
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Amanda Olivier
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Mohamed Farah
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Dawn Littlefield
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Supriya Behl
- Department of Pediatric Research, Mayo Clinic, Rochester, MN, United States
| | - Sohan Punia
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Reona Sakemura
- Department of Hematology Research, Mayo Clinic, Rochester, MN, United States
| | - Jack R. Hemsath
- Department of Infectious Diseases Research, Mayo Clinic, Rochester, MN, United States
| | - Andrew Norgan
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Elizabeth A. L. Enninga
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, United States,,Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Erica L. Johnson
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Rana Chakraborty
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States,,Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, United States,,Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, MN, United States,Correspondence: Rana Chakraborty
| |
Collapse
|
9
|
Megli CJ, Coyne CB. Infections at the maternal-fetal interface: an overview of pathogenesis and defence. Nat Rev Microbiol 2022; 20:67-82. [PMID: 34433930 PMCID: PMC8386341 DOI: 10.1038/s41579-021-00610-y] [Citation(s) in RCA: 216] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2021] [Indexed: 02/08/2023]
Abstract
Infections are a major threat to human reproductive health, and infections in pregnancy can cause prematurity or stillbirth, or can be vertically transmitted to the fetus leading to congenital infection and severe disease. The acronym 'TORCH' (Toxoplasma gondii, other, rubella virus, cytomegalovirus, herpes simplex virus) refers to pathogens directly associated with the development of congenital disease and includes diverse bacteria, viruses and parasites. The placenta restricts vertical transmission during pregnancy and has evolved robust mechanisms of microbial defence. However, microorganisms that cause congenital disease have likely evolved diverse mechanisms to bypass these defences. In this Review, we discuss how TORCH pathogens access the intra-amniotic space and overcome the placental defences that protect against microbial vertical transmission.
Collapse
Affiliation(s)
- Christina J Megli
- Division of Maternal-Fetal Medicine, Division of Reproductive Infectious Disease, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine and the Magee Womens Research Institute, Pittsburgh, PA, USA.
| | - Carolyn B Coyne
- Department of Molecular Genetics and Microbiology and the Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
10
|
Human cytomegalovirus inhibits the proliferation and invasion of extravillous cytotrophoblasts via Hippo-YAP pathway. Virol J 2021; 18:214. [PMID: 34717661 PMCID: PMC8557486 DOI: 10.1186/s12985-021-01681-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 10/13/2021] [Indexed: 11/30/2022] Open
Abstract
Background Human cytomegalovirus (HCMV) infection in utero is very common during pregnancy, which can lead to adverse outcomes in both pregnancy and progeny, but its pathogenesis has not been fully clarified. The decrease of extravillous cytotrophoblasts (EVT) invasion is an essential pathophysiological process of some pregnancy complications. Hippo-YAP signaling pathway plays an important role in regulating cell proliferation and apoptosis. However, whether YAP is involved in HCMV uterine infection remains to be studied. Methods The primary EVT was cultured and infected by the HCMV strain AD169 virus in vitro. Immunofluorescence staining of HCMVpp65 antigen was conducted afterward to confirm the establishment of an infection model. The optimal virus infection dose was determined by the EVT proliferation status in vitro. Real-time PCR was performed to examine the mRNA level of major genes involved in the Hippo pathway in EVT after HCMV infection. The effect of HCMV on the expression of YAP protein in EVT was evaluated by Immunofluorescence staining and Western blot. An in vitro cell invasion assay was carried out to analyze the influence of HCMV on EVT invasion. The changes of EVT invasion was accessed by establishing YAP silencing and over-expression models using YAP1 specific siRNA and plasmid pcDH. Results The optimal HCMV infection dose was 282.5TCID50/ml. Compared to the control group, the infection of HCMV significantly reduced the mRNA expression of Mst1, Mst2, SAV, Lats1, Lats2, Mob1, YAP1, TAZ, TEAD1-4 genes and YAP protein expression in the Hippo-YAP pathway. HCMV infection also decreased the EVT invasion. In non-infected EVT, the number of transmembrane EVT cells was significantly reduced when YAP1 gene was silenced, while it was significantly increased when YAP1 gene was over-expressed. In the HCMV-infected EVT, the number of transmembrane EVT cells significantly increased when over-expressed and eventually recovered to the level of NC. Conclusions HCMV may decrease EVT invasion by inhibiting the expression of mRNA and protein of YAP in the Hippo-YAP signaling pathway. HCMV eventually reduces the invasion ability of EVT by inhibiting multiple genes in the Hippo-YAP signaling pathway, especially inhibiting YAP which serves as the downstream effector.
Collapse
|
11
|
Butsabong T, Felippe M, Campagnolo P, Maringer K. The emerging role of perivascular cells (pericytes) in viral pathogenesis. J Gen Virol 2021; 102. [PMID: 34424156 PMCID: PMC8513640 DOI: 10.1099/jgv.0.001634] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Viruses may exploit the cardiovascular system to facilitate transmission or within-host dissemination, and the symptoms of many viral diseases stem at least in part from a loss of vascular integrity. The microvascular architecture is comprised of an endothelial cell barrier ensheathed by perivascular cells (pericytes). Pericytes are antigen-presenting cells (APCs) and play crucial roles in angiogenesis and the maintenance of microvascular integrity through complex reciprocal contact-mediated and paracrine crosstalk with endothelial cells. We here review the emerging ways that viruses interact with pericytes and pay consideration to how these interactions influence microvascular function and viral pathogenesis. Major outcomes of virus-pericyte interactions include vascular leakage or haemorrhage, organ tropism facilitated by barrier disruption, including viral penetration of the blood-brain barrier and placenta, as well as inflammatory, neurological, cognitive and developmental sequelae. The underlying pathogenic mechanisms may include direct infection of pericytes, pericyte modulation by secreted viral gene products and/or the dysregulation of paracrine signalling from or to pericytes. Viruses we cover include the herpesvirus human cytomegalovirus (HCMV, Human betaherpesvirus 5), the retrovirus human immunodeficiency virus (HIV; causative agent of acquired immunodeficiency syndrome, AIDS, and HIV-associated neurocognitive disorder, HAND), the flaviviruses dengue virus (DENV), Japanese encephalitis virus (JEV) and Zika virus (ZIKV), and the coronavirus severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2; causative agent of coronavirus disease 2019, COVID-19). We touch on promising pericyte-focussed therapies for treating the diseases caused by these important human pathogens, many of which are emerging viruses or are causing new or long-standing global pandemics.
Collapse
Affiliation(s)
- Teemapron Butsabong
- Department of Biochemical Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Mariana Felippe
- Department of Biochemical Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Paola Campagnolo
- Department of Biochemical Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Kevin Maringer
- The Pirbright Institute, Pirbright, Surrey, GU24 0NF, UK
| |
Collapse
|
12
|
Shuid AN, Jayusman PA, Shuid N, Ismail J, Kamal Nor N, Mohamed IN. Association between Viral Infections and Risk of Autistic Disorder: An Overview. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:2817. [PMID: 33802042 PMCID: PMC7999368 DOI: 10.3390/ijerph18062817] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/25/2021] [Accepted: 03/05/2021] [Indexed: 12/26/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition of the central nervous system (CNS) that presents with severe communication problems, impairment of social interactions, and stereotypic behaviours. Emerging studies indicate possible associations between viral infections and neurodegenerative and neurobehavioural conditions including autism. Viral infection during critical periods of early in utero neurodevelopment may lead to increased risk of autism in the offspring. This review is aimed at highlighting the association between viral infections, including viruses similar to COVID-19, and the aetiology of autism. A literature search was conducted using Pubmed, Ovid/Medline, and Google Scholar database. Relevant search terms included "rubella and autism", "cytomegalovirus and autism", "influenza virus and autism", "Zika virus and autism", "COVID-19 and autism". Based on the search terms, a total of 141 articles were obtained and studies on infants or children with congenital or perinatal viral infection and autistic behaviour were evaluated. The possible mechanisms by which viral infections could lead to autism include direct teratogenic effects and indirect effects of inflammation or maternal immune activation on the developing brain. Brain imaging studies have shown that the ensuing immune response from these viral infections could lead to disruption of the development of brain regions and structures. Hence, long-term follow up is necessary for infants whose mothers report an inflammatory event due to viral infection at any time during pregnancy to monitor for signs of autism. Research into the role of viral infection in the development of ASD may be one avenue of improving ASD outcomes in the future. Early screening and diagnosis to detect, and maybe even prevent ASD are essential to reduce the burden of this condition.
Collapse
Affiliation(s)
- Ahmad Naqib Shuid
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas 13200, Malaysia;
| | - Putri Ayu Jayusman
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Nazrun Shuid
- Department of Pharmacology, Faculty of Medicine, Universiti Teknologi MARA, Sg Buloh 47000, Malaysia
| | - Juriza Ismail
- Autism Research Group, Department of Pediatrics, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (J.I.); (N.K.N.)
| | - Norazlin Kamal Nor
- Autism Research Group, Department of Pediatrics, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (J.I.); (N.K.N.)
| | - Isa Naina Mohamed
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
13
|
Li Z, Li D, Rao Y, Wei L, Liu M, Zheng G, Yao Y, Hou X, Chen Y, Ruan XZ. SCAP knockout in SM22α-Cre mice induces defective angiogenesis in the placental labyrinth. Biomed Pharmacother 2021; 133:111011. [PMID: 33227706 DOI: 10.1016/j.biopha.2020.111011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
The placental labyrinth is important for the exchange of nutrients and gases between the mother and the embryo in mice. This interface contains cells of both trophoblast and allantoic mesodermal origin that together produce maternal blood sinuses and placental blood vessels. However, the molecular mechanisms that take place during process of placental labyrinth development, especially concerning fetal capillaries, are not well understood. SREBP cleavage-activating protein (SCAP), a membrane protein, is required for the synthesis of fatty acids and cholesterol. Recently, when we crossed the offspring of the cross between smooth muscle 22 alpha (SM22α)- Cre recombinase (Cre) mice and SCAPloxp/loxp mice to research the function of SCAP in vascular smooth muscle cells (VSMCs) during certain pathological processes, we found that there were no resultant SM22α-Cre-specific SCAP knockout (KO) pups (SM22α-Cre+SCAPflox/flox; hereafter referred to as SCAP KO). Through anatomic studies of these embryos and placentas, we found that SCAP KO resulted in defective placental vessels and abnormal fetal morphology. Further immunohistochemical and immunocytochemical analyses suggested that SCAP is knocked out in the pericytes of the placental labyrinth. Compared to wildtype mice, SCAP KO placentas had abnormal vasculature in the labyrinth and lower levels of angiogenesis. By using RNA-seq and western blotting, we found that the expression of some genes and proteins in SCAP KO placentas was changed, including those related to pericyte/endothelial interactions genes and angiogenesis. Our results suggest that the proper organizational structure of the placental labyrinth depends on SCAP expression in pericytes.
Collapse
Affiliation(s)
- Zhe Li
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Danyang Li
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yuhan Rao
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Li Wei
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Mihua Liu
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Guo Zheng
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yingcheng Yao
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xiaoli Hou
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yaxi Chen
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Xiong Z Ruan
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; John Moorhead Research Laboratory, Centre for Nephrology, University College London Medical School, Royal Free Campus, University College London, London, United Kingdom.
| |
Collapse
|
14
|
Serjilus A, Alcendor DJ. Unique method for human villous trophoblasts isolation from placental tissue explants. CLINICAL OBSTETRICS, GYNECOLOGY AND REPRODUCTIVE MEDICINE 2020; 6:319. [PMID: 33520289 PMCID: PMC7842261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Isolation of cytotrophoblasts from primary placental tissue may be costly and time consuming with variable results. In this paper, we provide a simple, affordable, and efficient method that may performed using common laboratory supplies to achieve consistent in vitro isolation of cytotrophoblasts from villous tissue. Trophoblast populations are identified based on morphology and phenotyping, which employs the timely extraction of villous nodes from the placenta prior to cultivation and isolation of nodal outgrowth by visual guidance for selective capture of cytotrophoblast populations and subculture. This method allows for the isolation of cytotrophoblasts free of contamination with other placental cell types. Isolated cells stain positive for the specific cytotrophoblast biomarker cytokeratin 7 and Human Chorionic Gonadotropin (HCG). Subcultured cells grow to confluency to establish monolayers that may be passaged in culture and later used to develop primary syncytiotrophoblasts over time. These primary cytotrophoblast populations may be employed using in in vitro placenta-on-a chip models to better understand placental cell biology and function, as well as physiological responses after exposure to toxicants, and infectious agents. This technique may be modified for selective isolation of specific cell types within different tissues from multiple organ systems.
Collapse
Affiliation(s)
- Ashley Serjilus
- Department of Obstetrics and Gynecology, School of Medicine, Meharry Medical College, USA
| | - Donald J Alcendor
- Center for AIDS Health Disparities Research, Department of Microbiology, Immunology, and Physiology, Meharry Medical College, School of Medicine, Nashville, TN, USA,Correspondence to: Donald J Alcendor, Associate Professor of Microbiology and Immunology, Center for AIDS Health Disparities Research, Meharry Medical College, 1005 Dr. D.B. Todd Jr. Blvd., Hubbard Hospital, 5th Floor, Rm. 5025, Nashville, TN 37208, USA,
| |
Collapse
|
15
|
Lesteberg KE, Fader DS, Beckham JD. Pregnancy Alters Innate and Adaptive Immune Responses to Zika Virus Infection in the Reproductive Tract. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:3107-3121. [PMID: 33127823 PMCID: PMC7686295 DOI: 10.4049/jimmunol.2000882] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/02/2020] [Indexed: 12/15/2022]
Abstract
Recent outbreaks of Zika virus (ZIKV) have been associated with birth defects, including microcephaly and neurologic impairment. However, the mechanisms that confer potential susceptibility to ZIKV during pregnancy remain unclear. We hypothesized that poor outcomes from ZIKV infection during pregnancy are due in part to pregnancy-induced alteration of innate immune cell frequencies and cytokine expression. To examine the impact of pregnancy on innate immune responses, we inoculated immunocompetent pregnant and nonpregnant female C57BL/6 mice with 5 × 105 focus-forming units of ZIKV intravaginally. Innate immune cell frequencies and cytokine expression were measured by flow cytometry at day 3 postinfection. Compared with nonpregnant mice, pregnant mice exhibited higher frequencies of uterine macrophages (CD68+) and CD11c+ CD103+ and CD11c+ CD11b+ dendritic cells. Additionally, ZIKV-infected pregnant mice had lower frequencies of CD45+ IL-12+ and CD11b+ IL-12+ cells in the uterus and spleen. Next, we measured the frequencies of Ag-experienced CD4 (CD4+ CD11a+ CD49d+) and CD8 (CD8lo CD11ahi) T cells at day 10 postinfection to determine the impact of pregnancy-associated changes in innate cellular IL-12 responses on the adaptive immune response. We found that pregnant mice had lower frequencies of uterine Ag-experienced CD4 T cells and ZIKV-infected pregnant mice had lower frequencies of uterine Ag-experienced CD8 T cells compared with ZIKV-infected nonpregnant mice. These data show that pregnancy results in altered innate and adaptive immune responses to ZIKV infection in the reproductive tract of mice and that pregnancy-associated immune modulation may play an important role in the severity of acute ZIKV infection.
Collapse
Affiliation(s)
- Kelsey E Lesteberg
- Department of Medicine, Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, CO 80045
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045
| | - Dana S Fader
- Department of Medicine, Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, CO 80045
| | - J David Beckham
- Department of Medicine, Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, CO 80045;
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO 80045; and
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045
| |
Collapse
|
16
|
Eastman AJ, Vrana EN, Grimaldo MT, Jones AD, Rogers LM, Alcendor DJ, Aronoff DM. Cytotrophoblasts suppress macrophage-mediated inflammation through a contact-dependent mechanism. Am J Reprod Immunol 2020; 85:e13352. [PMID: 32969101 DOI: 10.1111/aji.13352] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 08/31/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023] Open
Abstract
PROBLEM Gestational membrane (GM) infection provokes inflammation and can result in preterm prelabor rupture of membranes (PPROM). The choriodecidual layer of the GM includes decidual stromal cells (DSC), cytotrophoblasts (CTB), and macrophages (Mφ). Our laboratory has previously shown that DSCs suppress Mφ TNF-α production through secreted prostaglandin E2 . We hypothesized that CTBs would also inhibit Mφ cytokine expression through secreted mediators. METHOD OF STUDY THP.1 Mφ-like cells with an NF-κB reporter construct or human blood monocyte-derived Mφ were co-cultured with the Jeg3 CTB cell line or primary human CTBs and challenged with group B streptococcus (GBS) or Toll-like receptor (TLR) agonists. Conditioned medium generated from CTB cultures was applied to Mφ cultures before infection or treatment. Alternatively, CTBs were co-incubated with, but physically separated from, Mφ and GBS or TLR-stimulated. NF-κB was assessed via alkaline phosphatase assay, and proinflammatory mediators were assessed by qRT-PCR and ELISA. RESULTS CTBs suppressed GBS- or TLR-stimulated Mφ NF-κB activity, and TNF-α and MMP9 production. Direct physical contact between CTBs and Mφ was required for full immunosuppression. Immunosuppression could be overcome by increasing the ratio of Mφ to CTB. CONCLUSIONS CTBs limit Mφ NF-κB activation and production of TNF-α and MMP9 through an as-yet unknown, cell-to-cell contact-mediated mechanism. This suppression is distinct from the PGE2 -mediated Mφ TNF-α suppression by DSC, suggesting that DSCs and CTBs regulate Mφ inflammation through distinct mechanisms. How Mφ integrates these signals in an intact GM will be paramount to determining causes and prevention of PPROM.
Collapse
Affiliation(s)
- Alison J Eastman
- Division of Infectious Disease, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Erin N Vrana
- Vanderbilt University Medical School, Vanderbilt University, Nashville, TN, USA
| | - Maria T Grimaldo
- Texas A&M University, College of Agriculture and Life Sciences, College Station, TX, USA
| | - Amanda D Jones
- Division of Infectious Disease, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lisa M Rogers
- Division of Infectious Disease, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - David M Aronoff
- Division of Infectious Disease, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Pathology, Microbiology and Immunology, Department of Obstetrics and Gynecology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
17
|
Pique-Regi R, Romero R, Tarca AL, Luca F, Xu Y, Alazizi A, Leng Y, Hsu CD, Gomez-Lopez N. Does the human placenta express the canonical cell entry mediators for SARS-CoV-2? eLife 2020; 9:e58716. [PMID: 32662421 PMCID: PMC7367681 DOI: 10.7554/elife.58716] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
The pandemic of coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has affected more than 10 million people, including pregnant women. To date, no consistent evidence for the vertical transmission of SARS-CoV-2 exists. The novel coronavirus canonically utilizes the angiotensin-converting enzyme 2 (ACE2) receptor and the serine protease TMPRSS2 for cell entry. Herein, building upon our previous single-cell study (Pique-Regi et al., 2019), another study, and new single-cell/nuclei RNA-sequencing data, we investigated the expression of ACE2 and TMPRSS2 throughout pregnancy in the placenta as well as in third-trimester chorioamniotic membranes. We report that co-transcription of ACE2 and TMPRSS2 is negligible in the placenta, thus not a likely path of vertical transmission for SARS-CoV-2. By contrast, receptors for Zika virus and cytomegalovirus, which cause congenital infections, are highly expressed by placental cell types. These data show that the placenta minimally expresses the canonical cell-entry mediators for SARS-CoV-2.
Collapse
Affiliation(s)
- Roger Pique-Regi
- 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 ServicesDetroitUnited States
- Center for Molecular Medicine and Genetics, Wayne State University School of MedicineDetroitUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
| | - 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 ServicesDetroitUnited States
- Center for Molecular Medicine and Genetics, Wayne State University School of MedicineDetroitUnited States
- Department of Obstetrics and Gynecology, University of MichiganAnn ArborUnited States
- Department of Epidemiology and Biostatistics, Michigan State UniversityEast LansingUnited States
- Detroit Medical CenterDetroitUnited States
- Department of Obstetrics and Gynecology, Florida International UniversityMiamiUnited States
| | - Adi L Tarca
- 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 ServicesDetroitUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
- Department of Computer Science, Wayne State University College of EngineeringDetroitUnited States
| | - Francesca Luca
- Center for Molecular Medicine and Genetics, Wayne State University School of MedicineDetroitUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
| | - 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 ServicesDetroitUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
| | - Adnan Alazizi
- Center for Molecular Medicine and Genetics, Wayne State University School of MedicineDetroitUnited States
| | - Yaozhu Leng
- 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 ServicesDetroitUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
| | - Chaur-Dong Hsu
- 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 ServicesDetroitUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
- Department of Physiology, Wayne State University School of MedicineDetroitUnited States
| | - 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 ServicesDetroitUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of MedicineDetroitUnited States
| |
Collapse
|
18
|
Alcendor DJ. Human Vascular Pericytes and Cytomegalovirus Pathobiology. Int J Mol Sci 2019; 20:E1456. [PMID: 30909422 PMCID: PMC6471229 DOI: 10.3390/ijms20061456] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 03/01/2019] [Accepted: 03/20/2019] [Indexed: 12/11/2022] Open
Abstract
Pericytes are multipotent cells of the vascular system with cytoplasmic extensions proximal to endothelial cells that occur along the abluminal surface of the endothelium. The interactions between endothelial cells and pericytes are essential for proper microvascular formation, development, stabilization, and maintenance. Pericytes are essential for the regulation of paracellular flow between cells, transendothelial fluid transport, angiogenesis, and vascular immunosurveillance. They also influence the chemical composition of the surrounding microenvironment to protect endothelial cells from potential harm. Dysregulation or loss of pericyte function can result in microvascular instability and pathological consequences. Human pericytes have been shown to be targets for human cytomegalovirus (HCMV) infection and lytic replication that likely contribute to vascular inflammation. This review focuses on human vascular pericytes and their permissiveness for HCMV infection. It also discusses their implication in pathogenesis in the blood⁻brain barrier (BBB), the inner blood⁻retinal barrier (IBRB), the placenta⁻blood barrier, and the renal glomerulus as well as their potential role in subclinical vascular disease.
Collapse
Affiliation(s)
- Donald J Alcendor
- Center for AIDS Health Disparities Research, Department of Microbiology, Immunology, and Physiology, School of Medicine, Meharry Medical College, 1005 Dr. D.B. Todd Jr. Blvd., Hubbard Hospital, 5th Floor, Rm. 5025, Nashville, TN 37208, USA.
| |
Collapse
|
19
|
Barreto RSN, Romagnolli P, Cereta AD, Coimbra-Campos LMC, Birbrair A, Miglino MA. Pericytes in the Placenta: Role in Placental Development and Homeostasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1122:125-151. [PMID: 30937867 DOI: 10.1007/978-3-030-11093-2_8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The placenta is the most variable organ, in terms of structure, among the species. Besides it, all placental types have the same function: production of viable offspring, independent of pregnancy length, litter number, or invasion level. The angiogenesis is a central mechanism for placental functionality, due to proper maternal-fetal communication and exchanges. Much is known about the vasculature structure, but little is known about vasculature development and cellular interactions. Pericytes are perivascular cells that were described to control vasculature stability and permeability. Nowadays there are several new functions discovered, such as lymphocyte modulation and activation, macrophage-like phagocytic properties, tissue regenerative and repair processes, and also the ability to modulate stem cells, majorly the hematopoietic. In parallel, placental tissues are known to be a particularly immune microenvironment and a rich stem cell niche. The pericyte function plethora could be similar in the placental microenvironment and could have a central role in placental development and homeostasis.
Collapse
Affiliation(s)
- Rodrigo S N Barreto
- School of Veterinary Medicine and Animal Sciences, University of São Paulo, Butantã, Sao Paulo, Brazil
| | - Patricia Romagnolli
- School of Veterinary Medicine and Animal Sciences, University of São Paulo, Butantã, Sao Paulo, Brazil
| | - Andressa Daronco Cereta
- School of Veterinary Medicine and Animal Sciences, University of São Paulo, Butantã, Sao Paulo, Brazil
| | - Leda M C Coimbra-Campos
- Department of Pathology, Federal University of Minas Gerais, Pampulha, Belo Horizonte, Brazil
| | - Alexander Birbrair
- Department of Radiology, Columbia University Medical Center, New York, NY, USA.,Department of Pathology, Federal University of Minas Gerais, Pampulha, Belo Horizonte, Brazil
| | - Maria Angelica Miglino
- School of Veterinary Medicine and Animal Sciences, University of São Paulo, Butantã, Sao Paulo, Brazil.
| |
Collapse
|
20
|
Franke N, Bette M, Marquardt A, Briese T, Lipkin WI, Kurz C, Ehrenreich J, Mack E, Baying B, Beneš V, Rodepeter FR, Neff A, Teymoortash A, Eivazi B, Geisthoff U, Stuck BA, Bakowsky U, Mandic R. Virome Analysis Reveals No Association of Head and Neck Vascular Anomalies with an Active Viral Infection. In Vivo 2018; 32:1323-1331. [PMID: 30348684 DOI: 10.21873/invivo.11382] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 09/20/2018] [Accepted: 09/28/2018] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM Vascular anomalies encompass different vascular malformations [arteriovenous (AVM), lymphatic (LM), venous lymphatic (VLM), venous (VM)] and vascular tumors such as hemangiomas (HA). The pathogenesis of vascular anomalies is still poorly understood. Viral infection was speculated as a possible underlying cause. MATERIALS AND METHODS A total of 13 human vascular anomalies and three human skin control tissues were used for viral analysis. RNA derived from AVM (n=4) and normal skin control (n=3) tissues was evaluated by RNA sequencing. The Virome Capture Sequencing Platform for Vertebrate Viruses (VirCapSeq-VERT) was deployed on 10 tissues with vascular anomalies (2×AVM, 1×HA, 1×LM, 2×VLM, 4×VM). RESULTS RNA sequencing did not show any correlation of AVM with viral infection. By deploying VirCapSeq-VERT, no consistent viral association was seen in the tested tissues. CONCLUSION The analysis does not point to the presence of an active viral infection in vascular anomalies. However, transient earlier viral infections, e.g. during pregnancy, cannot be excluded with this approach.
Collapse
Affiliation(s)
- Nora Franke
- Department of Otolaryngology/Head and Neck Surgery, University Hospital Giessen and Marburg, Marburg, Germany
| | - Michael Bette
- Institute of Anatomy and Cell Biology, Philipps University, Marburg, Germany
| | - André Marquardt
- Department of Hematology, Oncology and Immunology, University Hospital Giessen and Marburg, Marburg, Germany
| | - Thomas Briese
- Center for Infection and Immunity, Columbia University, New York City, NY, U.S.A
| | - W Ian Lipkin
- Center for Infection and Immunity, Columbia University, New York City, NY, U.S.A
| | - Christopher Kurz
- Department of Otolaryngology/Head and Neck Surgery, University Hospital Giessen and Marburg, Marburg, Germany
| | - Jovine Ehrenreich
- Department of Otolaryngology/Head and Neck Surgery, University Hospital Giessen and Marburg, Marburg, Germany
| | - Elisabeth Mack
- Department of Hematology, Oncology and Immunology, University Hospital Giessen and Marburg, Marburg, Germany
| | - Bianka Baying
- Genomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Vladimir Beneš
- Genomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Andreas Neff
- Department of Oral and Maxillofacial Surgery, University Hospital Giessen and Marburg, Marburg, Germany
| | - Afshin Teymoortash
- Department of Otolaryngology/Head and Neck Surgery, University Hospital Giessen and Marburg, Marburg, Germany
| | - Behfar Eivazi
- Department of Otolaryngology/Head and Neck Surgery, University Hospital Giessen and Marburg, Marburg, Germany
| | - Urban Geisthoff
- Department of Otolaryngology/Head and Neck Surgery, University Hospital Giessen and Marburg, Marburg, Germany
| | - Boris A Stuck
- Department of Otolaryngology/Head and Neck Surgery, University Hospital Giessen and Marburg, Marburg, Germany
| | - Udo Bakowsky
- Department of Pharmaceutical Technology and Biopharmacy, Philipps University, Marburg, Germany
| | - Robert Mandic
- Department of Otolaryngology/Head and Neck Surgery, University Hospital Giessen and Marburg, Marburg, Germany
| |
Collapse
|
21
|
Popik W, Correa H, Khatua A, Aronoff DM, Alcendor DJ. Mesangial cells, specialized renal pericytes and cytomegalovirus infectivity: Implications for HCMV pathology in the glomerular vascular unit and post-transplant renal disease. ACTA ACUST UNITED AC 2018; 5. [PMID: 29977613 PMCID: PMC6027753 DOI: 10.15761/jts.1000248] [Citation(s) in RCA: 7] [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]
Abstract
Background Human Cytomegalovirus (HCMV) infection is problematic after kidney transplantation. Human mesangial cells along with human glomerular endothelial cells and podocytes constitute the renal glomerular vascular unit (GVU). HCMV infection of the GVU is poorly understood. Methods GVU cells infectivity was analysed by microscopy and immunofluorescence. Cytokines profiles were measured by Luminex assays. Renal tissue analysis for HCMV infection was performed by immunohistochemistry. Results Mesangial cells and glomerular endothelial cells but not podocytes were permissive for both lab adapted and clinical strains of HCMV. Luminex analysis of cytokines expressed by mesangial cells exposed to the SBCMV clinical strain was examined. A Tricell infection model of the GVU maintains >90% viability with a unique cytokine profile. Finally, we show αSMA stained mesangial cells permissive for HCMV in renal tissue from a transplant patient. Conclusions HCMV infection of mesangial cells induces angiogenic and proinflammatory cytokines that could contribute to glomerular inflammation.
Collapse
Affiliation(s)
- Waldemar Popik
- Department of Internal Medicine and 4Department of Microbiology and Immunology, Center for AIDS Health Disparities Research, Meharry Medical College, School of Medicine, 1005 Dr. D.B. Todd Jr. Blvd., Nashville, Tennessee 37208-3599 USA
| | - Hernan Correa
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee, USA
| | - Atanu Khatua
- Meharry Medical College, School of Medicine, Centre for AIDS Health Disparities Research, 1005 Dr. D.B. Todd Jr. Blvd., Nashville, Tennessee 37208-3599, USA
| | - David M Aronoff
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee, USA.,Division of Infectious Diseases, Department of Medicine, and Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Centre, Nashville, Tennessee 37232, USA
| | - Donald J Alcendor
- Meharry Medical College, School of Medicine, Centre for AIDS Health Disparities Research, 1005 Dr. D.B. Todd Jr. Blvd., Nashville, Tennessee 37208-3599, USA
| |
Collapse
|
22
|
Slawinski BL, Talge N, Ingersoll B, Smith A, Glazier A, Kerver J, Paneth N, Racicot K. Maternal cytomegalovirus sero-positivity and autism symptoms in children. Am J Reprod Immunol 2018; 79:e12840. [PMID: 29520885 PMCID: PMC5978736 DOI: 10.1111/aji.12840] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 02/10/2018] [Indexed: 12/28/2022] Open
Abstract
PROBLEM Autism spectrum disorder (ASD) is one of the most commonly diagnosed neurodevelopmental disorders in the United States. While ASD can be significantly influenced by genetics, prenatal exposure to maternal infections has also been implicated in conferring risk. Despite this, the effects of several important maternal pathogens, such as cytomegalovirus (CMV) and herpes simplex virus 2 (HSV2), remain unknown. METHOD OF STUDY We tested whether maternal CMV and/or HSV2 sero-positivity was associated with ASD symptoms in children. ELISA was used to assay for CMV IgG and HSV2 IgG in serum from the mothers of 82 children whose ASD symptoms were assessed at 3-6 years of age using the Social Responsiveness Scale version 2 (SRS-2). RESULTS Associations between maternal viral serostatus and SRS-2 scores were estimated using linear regression with covariate adjustments. The children of mothers sero-positive for CMV, but not for HSV2, had SRS-2 scores 3.6-4.2 points higher, depending on the adjustment model, than sero-negative women, a significant finding, robust to several statistical adjustments. CONCLUSION Our results suggest that maternal CMV infections may influence ASD symptoms. These findings are being further evaluated in ongoing prospective studies with larger population samples.
Collapse
Affiliation(s)
- Brooke L. Slawinski
- Department of Psychology, College of Social Sciences, Michigan State University, East Lansing, MI
| | - Nicole Talge
- Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Brooke Ingersoll
- Department of Psychology, College of Social Sciences, Michigan State University, East Lansing, MI
| | - Arianna Smith
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI
| | - Alicynne Glazier
- Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Jean Kerver
- Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Nigel Paneth
- Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Karen Racicot
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI
| |
Collapse
|