Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Viruses ; 14(5)2022 04 20.
Article in English | MEDLINE | ID: mdl-35632593

ABSTRACT

Zika virus (ZIKV) is a mosquito-borne RNA virus belonging to the Flavivirus genus of the Flaviviridae family. During the 60 years following its discovery in 1947, ZIKV caused little concern for public health as the associated infection was reported as mostly asymptomatic or inducing mild symptoms. However, since 2013, severe neurological symptoms have been associated with ZIKV infection, compelling the World Health Organization to declare a Public Health Emergency of International Concern. Among those symptoms, neurological birth defects may affect children born to mothers infected during pregnancy. Additionally, during the past 8 years, ZIKV transmission through breastfeeding has repeatedly been suggested in epidemiological studies and demonstrated on a mouse model by our team. To better understand the biological factors controlling ZIKV transmission through breastfeeding, we investigated the nature of the viral entities excreted in the breast milk of infected dams and evaluated viral transmission to breastfed pups. We show that both cell-free and cell-associated virus is excreted into breast milk and that ZIKV is efficiently transmitted to the breastfed pups. Additionally, we studied murine breast milk cell types, and identified a majority of mammary luminal cells. Finally, we investigated the effect on ZIKV infectivity of several breast milk components that are antiviral against different viruses such as lactoferrin (LF) and lactalbumin (LA), or free fatty acids (FFA). We showed no effect of LF and LA, whereas FFA inactivated the virus. These results bring new insight concerning the mechanisms of ZIKV transmission during breastfeeding and identify biological factors modulating it. These elements should be considered in risk assessment of ZIKV mother-to-child transmission.


Subject(s)
Zika Virus Infection , Zika Virus , Animals , Antiviral Agents/pharmacology , Biological Factors/pharmacology , Female , Humans , Infectious Disease Transmission, Vertical , Mice , Milk, Human , Pregnancy , Satellite Viruses , Zika Virus/genetics
2.
Front Microbiol ; 12: 746589, 2021.
Article in English | MEDLINE | ID: mdl-34616388

ABSTRACT

Zika virus (ZIKV) infection has been associated with a series of neurological pathologies. In patients with ZIKV-induced neurological disorders, the virus is detectable in the central nervous system. Thus, ZIKV is capable of neuroinvasion, presumably through infection of the endothelial cells that constitute the blood-brain barrier (BBB). We demonstrate that susceptibility of BBB endothelial cells to ZIKV infection is modulated by the expression of tight-junction protein claudin-7 (CLDN7). Downregulation of CLDN7 reduced viral RNA yield, viral protein production, and release of infectious viral particles in several endothelial cell types, but not in epithelial cells, indicating that CLDN7 implication in viral infection is cell-type specific. The proviral activity of CLDN7 in endothelial cells is ZIKV-specific since related flaviviruses were not affected by CLDN7 downregulation. Together, our data suggest that CLDN7 facilitates ZIKV infection in endothelial cells at a post-internalization stage and prior to RNA production. Our work contributes to a better understanding of the mechanisms exploited by ZIKV to efficiently infect and replicate in endothelial cells and thus of its ability to cross the BBB.

3.
Viruses ; 13(7)2021 07 07.
Article in English | MEDLINE | ID: mdl-34372518

ABSTRACT

Most viruses use several entry sites and modes of transmission to infect their host (parenteral, sexual, respiratory, oro-fecal, transplacental, transcutaneous, etc.). Some of them are known to be essentially transmitted via arthropod bites (mosquitoes, ticks, phlebotomes, sandflies, etc.), and are thus named arthropod-borne viruses, or arboviruses. During the last decades, several arboviruses have emerged or re-emerged in different countries in the form of notable outbreaks, resulting in a growing interest from scientific and medical communities as well as an increase in epidemiological studies. These studies have highlighted the existence of other modes of transmission. Among them, mother-to-child transmission (MTCT) during breastfeeding was highlighted for the vaccine strain of yellow fever virus (YFV) and Zika virus (ZIKV), and suggested for other arboviruses such as Chikungunya virus (CHIKV), dengue virus (DENV), and West Nile virus (WNV). In this review, we summarize all epidemiological and clinical clues that suggest the existence of breastfeeding as a neglected route for MTCT of arboviruses and we decipher some of the mechanisms that chronologically occur during MTCT via breastfeeding by focusing on ZIKV transmission process.


Subject(s)
Arbovirus Infections/epidemiology , Arbovirus Infections/transmission , Arboviruses/pathogenicity , Breast Feeding , Infectious Disease Transmission, Vertical , Milk, Human/virology , Animals , Arboviruses/classification , Chikungunya Fever/transmission , Chikungunya Fever/virology , Colostrum/virology , Culicidae/virology , Dengue/transmission , Dengue/virology , Disease Outbreaks , Female , Humans , West Nile Fever/transmission , West Nile Fever/virology , Zika Virus Infection/transmission , Zika Virus Infection/virology
4.
Pediatr Allergy Immunol ; 32(5): 835-842, 2021 07.
Article in English | MEDLINE | ID: mdl-33594740

ABSTRACT

As breastfeeding is of utmost importance for child development and survival, identifying whether breast milk is a route of transmission for human viruses is critical. Based on the principle of Koch's postulate, we propose an analytical framework to determine the plausibility of viral transmission by breast milk. This framework is based on five criteria: viral infection in children receiving breast milk from infected mothers; the presence of virus, viral antigen, or viral genome in the breast milk of infected mothers; the evidence for the virus in breast milk being infectious; the attempts to rule out other transmission modalities; and the reproduction of viral transmission by oral inoculation in an animal model. We searched for evidence in published reports to determine whether the 5 criteria are fulfilled for 16 human viruses that are suspected to be transmissible by breast milk. We considered breast milk transmission is proven if all 5 criteria are fulfilled, as probable if 4 of the 5 criteria are met, as possible if 3 of the 5 criteria are fulfilled, and as unlikely if less than 3 criteria are met. Only five viruses have proven transmission through breast milk: human T-cell lymphotropic virus 1, human immunodeficiency virus, human cytomegalovirus, dengue virus, and Zika virus. The other 11 viruses fulfilled some but not all criteria and were categorized accordingly. Our framework analysis is useful for guiding public health recommendations and for identifying knowledge gaps amenable to original experiments.


Subject(s)
HIV Infections , Virus Diseases , Zika Virus Infection , Zika Virus , Animals , Breast Feeding , Female , Humans , Infectious Disease Transmission, Vertical , Milk, Human
5.
Front Microbiol ; 11: 524678, 2020.
Article in English | MEDLINE | ID: mdl-33193119

ABSTRACT

Zika virus (ZIKV) belongs to the Flavivirus genus in the Flaviviridae family. Mainly transmitted via mosquito bites (Aedes aegypti, Aedes albopictus), ZIKV has been classified in the large category of arthropod-borne viruses, or arboviruses. However, during the past two outbreaks in French Polynesia (2013-2014) and Latin America (2015-2016), several cases of ZIKV human-to-human transmission were reported, either vertically via transplacental route but also horizontally after sexual intercourse. Interestingly, high viral burdens were detected in the colostrum and breast milk of infected women and mother-to-child transmission of ZIKV during breastfeeding was recently highlighted. In a previous study, we highlighted the implication of the mammary epithelium (blood-milk barrier) in ZIKV infectious particles excretion in breast milk. However, mechanisms of their further transmissibility to the newborn via oral route through contaminated breast milk remain unknown. In this study, we provide the first experimental proof-of-concept of the existence of the breastfeeding as a route for mother-to-child transmission of ZIKV and characterized the neonatal oral transmission in a well-established mouse model of ZIKV infection. From a mechanistical point-of-view, we demonstrated for the first time that ZIKV was able to infect and cross an in vitro model of tight human intestinal epithelium without altering its barrier integrity, permitting us to consider the gut as an entry site for ZIKV after oral exposure. By combining in vitro and in vivo experiments, this study strengthens the plausibility of mother-to-child transmission of ZIKV during breastfeeding and helps to better characterize underlying mechanisms, such as the crossing of the newborn intestinal epithelium by ZIKV. As a consequence, these data could serve as a basis for a reflection about the implementation of measures to prevent ZIKV transmission, while keeping in mind breastfeeding-associated benefits.

6.
PLoS Negl Trop Dis ; 14(8): e0008282, 2020 08.
Article in English | MEDLINE | ID: mdl-32817655

ABSTRACT

Muscle cells are potential targets of many arboviruses, such as Ross River, Dengue, Sindbis, and chikungunya viruses, that may be involved in the physiopathological course of the infection. During the recent outbreak of Zika virus (ZIKV), myalgia was one of the most frequently reported symptoms. We investigated the susceptibility of human muscle cells to ZIKV infection. Using an in vitro model of human primary myoblasts that can be differentiated into myotubes, we found that myoblasts can be productively infected by ZIKV. In contrast, myotubes were shown to be resistant to ZIKV infection, suggesting a differentiation-dependent susceptibility. Infection was accompanied by a caspase-independent cytopathic effect, associated with paraptosis-like cytoplasmic vacuolization. Proteomic profiling was performed 24h and 48h post-infection in cells infected with two different isolates. Proteome changes indicate that ZIKV infection induces an upregulation of proteins involved in the activation of the Interferon type I pathway, and a downregulation of protein synthesis. This work constitutes the first observation of primary human muscle cells susceptibility to ZIKV infection, and differentiation-dependent restriction of infection from myoblasts to myotubes. Since myoblasts constitute the reservoir of stem cells involved in reparation/regeneration in muscle tissue, the infection of muscle cells and the viral-induced alterations observed here could have consequences in ZIKV infection pathogenesis.


Subject(s)
Cell Differentiation , Muscle Cells/metabolism , Muscle Cells/virology , Proteomics , Zika Virus Infection , Cell Death , Cell Line , Cytopathogenic Effect, Viral , Disease Susceptibility , Host-Pathogen Interactions , Humans , Interferon Type I/metabolism , Muscle Cells/pathology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/virology , Myoblasts/metabolism , Myoblasts/virology , Proteins/metabolism , Stem Cells , Virus Replication , Zika Virus/pathogenicity , Zika Virus Infection/pathology , Zika Virus Infection/virology
7.
Viruses ; 12(6)2020 06 05.
Article in English | MEDLINE | ID: mdl-32516914

ABSTRACT

Infections due to arboviruses (arthropod-borne viruses) have dramatically increased worldwide during the last few years. In humans, symptoms associated with acute infection of most arboviruses are often described as "dengue-like syndrome", including fever, rash, conjunctivitis, arthralgia, and muscular symptoms such as myalgia, myositis, or rhabdomyolysis. In some cases, muscular symptoms may persist over months, especially following flavivirus and alphavirus infections. However, in humans the cellular targets of infection in muscle have been rarely identified. Animal models provide insights to elucidate pathological mechanisms through studying viral tropism, viral-induced inflammation, or potential viral persistence in the muscle compartment. The tropism of arboviruses for muscle cells as well as the viral-induced cytopathic effect and cellular alterations can be confirmed in vitro using cellular models. This review describes the link between muscle alterations and arbovirus infection, and the underlying mechanisms.


Subject(s)
Arbovirus Infections/virology , Arboviruses/physiology , Muscular Diseases/virology , Animals , Arbovirus Infections/pathology , Arboviruses/genetics , Cytopathogenic Effect, Viral , Humans , Muscles/virology , Muscular Diseases/pathology
8.
Viruses ; 11(10)2019 10 15.
Article in English | MEDLINE | ID: mdl-31619008

ABSTRACT

Zika virus (ZIKV) belongs to the large category of arboviruses. Surprisingly, several human-to-human transmissions of ZIKV have been notified, either following sexual intercourse or from the mother to fetus during pregnancy. Importantly, high viral loads have been detected in the human breast milk of infected mothers, and the existence of breastfeeding as a new mode of mother-to-child transmission of ZIKV was recently hypothesized. However, the maternal origin of infectious particles in breast milk is currently unknown. Here, we show that ZIKV disseminates to the mammary glands of infected mice after both systemic and local exposure with differential kinetics. Ex vivo, we demonstrate that primary human mammary epithelial cells were sensitive and permissive to ZIKV infection in this study. Moreover, by using in vitro models, we prove that mammary luminal- and myoepithelial-phenotype cell lines are both able to produce important virus progeny after ZIKV exposure. Our data suggest that the dissemination of ZIKV to the mammary glands and subsequent infection of the mammary epithelium could be one mechanism of viral excretion in human breast milk.


Subject(s)
Epithelial Cells/virology , Mammary Glands, Human/virology , Viral Tropism , Virus Replication , Zika Virus/growth & development , Animals , Cell Line , Female , Humans , Infectious Disease Transmission, Vertical , Mammary Glands, Human/cytology , Mice , Milk, Human/virology , Pregnancy , RNA, Viral , Viral Load , Zika Virus/genetics , Zika Virus/physiology
9.
J Vis Exp ; (146)2019 04 09.
Article in English | MEDLINE | ID: mdl-31033945

ABSTRACT

The early screening of nervous system medicines on a pertinent and reliable in cellulo BBB model for their penetration and their interaction with the barrier and the brain parenchyma is still an unmet need. To fill this gap, we designed a 2D in cellulo model, the BBB-Minibrain, by combining a polyester porous membrane culture insert human BBB model with a Minibrain formed by a tri-culture of human brain cells (neurons, astrocytes and microglial cells). The BBB-Minibrain allowed us to test the transport of a neuroprotective drug candidate (e.g., Neurovita), through the BBB, to determine the specific targeting of this molecule to neurons and to show that the neuroprotective property of the drug was preserved after the drug had crossed the BBB. We have also demonstrated that BBB-Minibrain constitutes an interesting model to detect the passage of virus particles across the endothelial cells barrier and to monitor the infection of the Minibrain by neuroinvasive virus particles. The BBB-Minibrain is a reliable system, easy to handle for researcher trained in cell culture technology and predictive of the brain cells phenotypes after treatment or insult. The interest of such in cellulo testing would be twofold: introducing derisking steps early in the drug development on the one hand and reducing the use of animal testing on the other hand.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , Models, Neurological , Neuroprotective Agents/metabolism , Animals , Astrocytes/physiology , Blood-Brain Barrier/physiology , Cells, Cultured , Endothelial Cells/physiology , Humans , Neurons/metabolism , Neuroprotective Agents/administration & dosage
10.
Influenza Other Respir Viruses ; 13(4): 398-406, 2019 07.
Article in English | MEDLINE | ID: mdl-29144593

ABSTRACT

BACKGROUND: Previous studies reported detection of influenza RNA in stools of patients with seasonal influenza infection. While this detection may have a clinical significance, other factors may influence the stool positivity for influenza viruses. OBJECTIVES: The objective of this study was to investigate demographical, clinical, and microbiological factors which could favor the presence of influenza viral RNA in the stools of patients with laboratory-confirmed influenza infection. METHODS: Acute respiratory infection (ARI) patients were enrolled by general practitioners (GP) during two winter seasons (2014-2016). Nasopharyngeal swabs, stool specimens, and clinical data were collected. Samples were tested for 12 respiratory pathogen groups (nasopharyngeal and stool specimens) and for 12 enteric pathogens (stool specimens). RESULTS: Among the 331 patients with ARI enrolled by GP, 114 (34.4%) presented influenza infection. Influenza RNA was detected in stool samples of 21% (24/114) of the 114 stool specimens analyzed. Hospitalization (adjusted odds ratio (aOR) = 7.8 (95% confidence interval (CI)) [1.7-33.7], P = .02), age between 45 and 64 years (aOR = 4.8 [1.7-14.5], P = .01), consumption of raw shellfish and/or mollusks (aOR = 16.7 [3.6-90.9], P = .00), and use of antibiotics (aOR = 6.4 [2.1-19.8], P = .006) or antiviral treatment (aOR = 7.4 [1.9-29], P = .01) were significantly associated with an increased odds of the detection of influenza RNA in stools. Among the 24 stool samples subjected to viral isolation, no one showed virus growth. CONCLUSIONS: These findings will be useful to studies investigating the dissemination route of influenza viruses to gastrointestinal tract.


Subject(s)
Feces/virology , General Practice , Influenza, Human/epidemiology , Orthomyxoviridae/isolation & purification , Respiratory Tract Infections/epidemiology , Adolescent , Adult , Aged , Child , Child, Preschool , Cross-Sectional Studies , Female , France/epidemiology , Humans , Infant , Infant, Newborn , Male , Middle Aged , Nasopharynx/virology , Orthomyxoviridae/genetics , Prospective Studies , RNA, Viral/analysis , Respiratory Tract Infections/virology , Risk Factors , Young Adult
11.
BMC Infect Dis ; 17(1): 729, 2017 11 22.
Article in English | MEDLINE | ID: mdl-29166867

ABSTRACT

BACKGROUND: Gastrointestinal (GI) symptoms, such as diarrhea, vomiting, abdominal pain and nausea are not an uncommon manifestation of an acute respiratory infection (ARI). We therefore evaluated clinical and microbiological factors associated with the presence of GI symptoms in patients consulting a general practitioner (GP) for ARI. METHODS: Nasopharyngeal swabs, stool specimens and clinical data from patients presenting to GPs with an ARI were prospectively collected during two winter seasons (2014-2016). Samples were tested by quantitative real-time PCR for 12 respiratory pathogen groups and for 12 enteric pathogens. RESULTS: Two hundred and four of 331 included patients (61.6%) were positive for at least one respiratory pathogen. Sixty-nine stools (20.8%) were positive for at least one pathogen (respiratory and/or enteric). GI symptoms were more likely declared in case of laboratory confirmed-enteric infection (adjusted odds ratio (aOR) = 3.2; 95% confidence interval [CI] [1.2-9.9]; p = 0.02) or human coronavirus (HCoV) infection (aOR = 2.7; [1.2-6.8]; p = 0.02). Consumption of antipyretic medication before the consultation seemed to reduce the risk of developing GI symptoms for patients with laboratory-confirmed influenza (aOR = 0.3; [0.1-0.6]; p = 0.002). CONCLUSIONS: The presence of GI symptoms in ARI patients could not be explained by the detection of respiratory pathogens in stools. However, the detection of enteric pathogens in stool samples could explained by the presence of GI symptoms in some of ARI cases. The biological mechanisms explaining the association between the presence of HCoVs in nasopharynx and GI symptoms need to be explored.


Subject(s)
Gastrointestinal Diseases/virology , Influenza, Human/etiology , Respiratory Tract Infections/complications , Adolescent , Adult , Aged , Child , Child, Preschool , Diarrhea/virology , Feces/virology , Female , France , Gastrointestinal Diseases/etiology , Gastrointestinal Diseases/microbiology , General Practice , Humans , Infant , Influenza, Human/virology , Male , Middle Aged , Nasopharynx/virology , Nausea/etiology , Nausea/virology , Prospective Studies , Real-Time Polymerase Chain Reaction , Respiratory Tract Infections/virology , Seasons
12.
Nat Commun ; 8: 15890, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28639618

ABSTRACT

The human T-lymphotropic virus type 1 (HTLV-1) is efficiently transmitted through cellular contacts. While the molecular mechanisms of viral cell-to-cell propagation have been extensively studied in vitro, those facilitating the encounter between infected and target cells remain unknown. In this study, we demonstrate that HTLV-1-infected CD4 T cells secrete a potent chemoattractant, leukotriene B4 (LTB4). LTB4 secretion is dependent on Tax-induced transactivation of the pla2g4c gene, which encodes the cytosolic phospholipase A2 gamma. Inhibition of LTB4 secretion or LTB4 receptor knockdown on target cells reduces T-cell recruitment, cellular contact formation and virus propagation in vitro. Finally, blocking the synthesis of LTB4 in a humanized mouse model of HTLV-1 infection significantly reduces proviral load. This results from a decrease in the number of infected clones while their expansion is not impaired. This study shows the critical role of LTB4 secretion in HTLV-1 transmission both in vitro and in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Human T-lymphotropic virus 1/pathogenicity , Leukotriene B4/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Gene Products, tax/genetics , Gene Products, tax/metabolism , Group IV Phospholipases A2/genetics , Group IV Phospholipases A2/metabolism , HTLV-I Infections/drug therapy , HTLV-I Infections/metabolism , HTLV-I Infections/virology , Host-Pathogen Interactions , Humans , Indoles/pharmacology , Infant, Newborn , Jurkat Cells , Lipoxygenase Inhibitors/pharmacology , Male , Mice, Mutant Strains , NF-kappa B/genetics , NF-kappa B/metabolism
13.
Virologie (Montrouge) ; 21(1): 11-18, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-31967563

ABSTRACT

HTLV-1 (Human T-cell Lymphotropic Virus Type 1) is a human retrovirus that infects around 10 million people worldwide. It can be transmitted by sexual contact, transfusion of contaminated blood, and from infected mother-to-child during prolonged breastfeeding. The latter involves viral crossing of the digestive tract. HTLV-1 is the etiological agent of both a lymphoproliferative malignancy, Adult T-cell leukemia/lymphoma, and a chronic inflammatory neuromyelopathy, the Tropical Spastic Paraparesis/HTLV-1 Associated Myelopathy (TSP/HAM). TSP/HAM is characterized by HTLV-1-infected lymphocyte infiltration in the central nervous system; these cells cross the blood-brain barrier, an anatomical barrier that normally isolates and protects the central nervous system from blood. In this context, the present review focuses on latest findings and opinions on the interactions of HTLV-1 with the intestinal barrier, as involved in mother-to-child viral transmission, and with the blood-brain barrier, as involved in TSP/HAM pathogenesis.

14.
J Virol ; 90(16): 7303-7312, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27252538

ABSTRACT

UNLABELLED: Human T-lymphotropic virus type 1 (HTLV-1) is the etiological agent of a slowly progressive neurodegenerative disease, HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). This disease develops upon infiltration of HTLV-1-infected lymphocytes into the central nervous system, mostly the thoracic spinal cord. The central nervous system is normally protected by a physiological structure called the blood-brain barrier (BBB), which consists primarily of a continuous endothelium with tight junctions. In this study, we investigated the role of activated leukocyte cell adhesion molecule (ALCAM/CD166), a member of the immunoglobulin superfamily, in the crossing of the BBB by HTLV-1-infected lymphocytes. We demonstrated that ALCAM is overexpressed on the surface of HTLV-1-infected lymphocytes, both in chronically infected cell lines and in primary infected CD4(+) T lymphocytes. ALCAM overexpression results from the activation of the canonical NF-κB pathway by the viral transactivator Tax. In contrast, staining of spinal cord sections of HAM/TSP patients showed that ALCAM expression is not altered on the BBB endothelium in the context of HTLV-1 infection. ALCAM blockade or downregulation of ALCAM levels significantly reduced the migration of HTLV-1-infected lymphocytes across a monolayer of human BBB endothelial cells. This study suggests a potential role for ALCAM in HAM/TSP pathogenesis. IMPORTANCE: Human T-lymphotropic virus type 1 (HTLV-1) is the etiological agent of a slowly progressive neurodegenerative disease, HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). This disease is the consequence of the infiltration of HTLV-1-infected lymphocytes into the central nervous system (CNS), mostly the thoracic spinal cord. The CNS is normally protected by a physiological structure called the blood-brain barrier (BBB), which consists primarily of a continuous endothelium with tight junctions. The mechanism of migration of lymphocytes into the CNS is unclear. Here, we show that the viral transactivator Tax increases activated leukocyte cell adhesion molecule (ALCAM/CD166) expression. This molecule facilitates the migration of lymphocytes across the BBB endothelium. Targeting this molecule could be of interest in preventing or reducing the development of HAM/TSP.


Subject(s)
Antigens, CD/metabolism , Blood-Brain Barrier , CD4-Positive T-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/virology , Cell Adhesion Molecules, Neuronal/metabolism , Cell Movement , Fetal Proteins/metabolism , Host-Pathogen Interactions , Human T-lymphotropic virus 1/physiology , CD4-Positive T-Lymphocytes/chemistry , Cell Line , Endothelial Cells/chemistry , Gene Products, tax/metabolism , Humans , NF-kappa B/metabolism
15.
Viruses ; 8(2)2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26848683

ABSTRACT

Human T-cell Lymphotropic Virus type 1 (HTLV-1) is a human retrovirus that infects at least 5-10 million people worldwide, and is the etiological agent of a lymphoproliferative malignancy; Adult T-cell Leukemia/Lymphoma (ATLL); and a chronic neuromyelopathy, HTLV-1 Associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP), as well as other inflammatory diseases such as infective dermatitis and uveitis. Besides sexual intercourse and intravenous transmission, HTLV-1 can also be transmitted from infected mother to child during prolonged breastfeeding. Some characteristics that are linked to mother-to-child transmission (MTCT) of HTLV-1, such as the role of proviral load, antibody titer of the infected mother, and duration of breastfeeding, have been elucidated; however, most of the mechanisms underlying HTLV-1 transmission during breast feeding remain largely unknown, such as the sites of infection and cellular targets as well as the role of milk factors. The present review focuses on the latest findings and current opinions and perspectives on MTCT of HTLV-1.


Subject(s)
HTLV-I Infections/transmission , Human T-lymphotropic virus 1/physiology , Adult , Female , HTLV-I Infections/epidemiology , HTLV-I Infections/virology , Human T-lymphotropic virus 1/genetics , Humans , Infant , Infectious Disease Transmission, Vertical , Male
16.
Vet Res ; 47: 9, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26743565

ABSTRACT

Sleeping disease in rainbow trout is characterized by an abnormal swimming behaviour of the fish which stay on their side at the bottom of the tanks. This sign is due to extensive necrosis and atrophy of red skeletal muscle induced by the sleeping disease virus (SDV), also called salmonid alphavirus 2. Infections of humans with arthritogenic alphaviruses, such as Chikungunya virus (CHIKV), are global causes of debilitating musculoskeletal diseases. The mechanisms by which the virus causes these pathologies are poorly understood due to the restrictive availability of animal models capable of reproducing the full spectrum of the disease. Nevertheless, it has been shown that CHIKV exhibits a particular tropism for muscle stem cells also known as satellite cells. Thus, SDV and its host constitute a relevant model to study in details the virus-induced muscle atrophy, the pathophysiological consequences of the infection of a particular cell-type in the skeletal muscle, and the regeneration of the muscle tissue in survivors together with the possible virus persistence. To study a putative SDV tropism for that particular cell type, we established an in vivo and ex vivo rainbow trout model of SDV-induced atrophy of the skeletal muscle. This experimental model allows reproducing the full panel of clinical signs observed during a natural infection since the transmission of the virus is arthropod-borne independent. The virus tropism in the muscle tissue was studied by immunohistochemistry together with the kinetics of the muscle atrophy, and the muscle regeneration post-infection was observed. In parallel, an ex vivo model of SDV infection of rainbow trout satellite cells was developed and virus replication and persistence in that particular cell type was followed up to 73 days post-infection. These results constitute the first observation of a specific SDV tropism for the muscle satellite cells.


Subject(s)
Alphavirus Infections/veterinary , Alphavirus/classification , Fish Diseases/virology , Oncorhynchus mykiss , Satellite Cells, Skeletal Muscle/virology , Alphavirus Infections/virology , Animals , Muscle, Skeletal/pathology , Muscular Atrophy/veterinary , Muscular Atrophy/virology , Regeneration
17.
Virol J ; 12: 215, 2015 Dec 12.
Article in English | MEDLINE | ID: mdl-26651485

ABSTRACT

This review provides for the first time an assessment of the current understanding about the occurrence and the clinical significance of gastrointestinal (GI) symptoms in influenza patients, and their correlation with the presence of human influenza viruses in stools of patients with confirmed influenza virus infection. Studies exploring how human influenza viruses spread to the patient's GI tract after a primary respiratory infection have been summarized. We conducted a systematic search of published peer-reviewed literature up to June 2015 with regard to the above-mentioned aspects, focusing on human influenza viruses (A(H1N1), A(H1N1)pdm09, A(H3N2), and B). Forty-four studies were included in this systematic review and meta-analysis. The pooled prevalence of any digestive symptoms ranged from 30.9% (95% CI, 9.8 to 57.5; I(2) = 97.5%) for A(H1N1)pdm09 to 2.8% (95% CI, 0.6 to 6.5; I(2) = 75.4%) for A(H1N1). The pooled prevalence of influenza viruses in stool was 20.6% (95% CI, 8.9 to 35.5; I(2) = 96.8%), but their correlation with GI symptoms has rarely been explored. The presence of viral RNA in stools because of haematogenous dissemination to organs via infected lymphocytes is likely, but the potential to cause direct intestinal infection and faecal-oral transmission warrants further investigation. This review highlights the gaps in our knowledge, and the high degree of uncertainty about the prevalence and significance of GI symptoms in patients with influenza and their correlation with viral RNA positivity in stool because of the high level of heterogeneity among studies.


Subject(s)
Feces/virology , Gastrointestinal Diseases/epidemiology , Gastrointestinal Diseases/etiology , Influenza, Human/complications , Influenza, Human/pathology , Orthomyxoviridae/isolation & purification , Gastrointestinal Diseases/pathology , Humans , Prevalence
18.
PLoS One ; 8(11): e79628, 2013.
Article in English | MEDLINE | ID: mdl-24223983

ABSTRACT

Besides the classical respiratory and systemic symptoms, unusual complications of influenza A infection in humans involve the skeletal muscles. Numerous cases of acute myopathy and/or rhabdomyolysis have been reported, particularly following the outbreak of pandemic influenza A(H1N1) in 2009. The pathogenesis of these influenza-associated myopathies (IAM) remains unkown, although the direct infection of muscle cells is suspected. Here, we studied the susceptibility of cultured human primary muscle cells to a 2009 pandemic and a 2008 seasonal influenza A(H1N1) isolate. Using cells from different donors, we found that differentiated muscle cells (i. e. myotubes) were highly susceptible to infection by both influenza A(H1N1) isolates, whereas undifferentiated cells (i. e. myoblasts) were partially resistant. The receptors for influenza viruses, α2-6 and α2-3 linked sialic acids, were detected on the surface of myotubes and myoblasts. Time line of viral nucleoprotein (NP) expression and nuclear export showed that the first steps of the viral replication cycle could take place in muscle cells. Infected myotubes and myoblasts exhibited budding virions and nuclear inclusions as observed by transmission electron microscopy and correlative light and electron microscopy. Myotubes, but not myoblasts, yielded infectious virus progeny that could further infect naive muscle cells after proteolytic treatment. Infection led to a cytopathic effect with the lysis of muscle cells, as characterized by the release of lactate dehydrogenase. The secretion of proinflammatory cytokines by muscle cells was not affected following infection. Our results are compatible with the hypothesis of a direct muscle infection causing rhabdomyolysis in IAM patients.


Subject(s)
Influenza A Virus, H1N1 Subtype/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/virology , Pandemics , Seasons , Cell Death , Cell Differentiation , Cell Proliferation , Humans , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/virology , Myoblasts/cytology , Myoblasts/virology , Receptors, Cell Surface/metabolism , Virus Replication
19.
J Clin Virol ; 57(1): 70-6, 2013 May.
Article in English | MEDLINE | ID: mdl-23375238

ABSTRACT

BACKGROUND: The Human T-cell Leukemia Virus type 1 (HTLV-1) is the causative agent of several inflammatory diseases, including HTLV-1-associated inflammatory myopathies (HAIM). Little is known about the virological and immunological characteristics of this viral disease. OBJECTIVES: To characterize the histological and virological features of HAIM patients, in order to better understand the pathogenetic mechanisms and unravel new biological markers of this disease. STUDY DESIGN: We conducted a retrospective study on 13 patients with HAIM, based on blood and muscle samples. We included blood samples from HTLV-1-infected individuals without myopathy as controls. Muscle biopsies were used for a broad immunohistological evaluation of tissue damage and inflammation, as well as identification of infected cells through in situ hybridization. DNA extracted from patients' PBMC was used to identify the virus genotype by sequencing and to assess the proviral load by quantitative PCR. Anti-viral antibodies in plasma samples were titrated by indirect immunofluorescence. RESULTS: Patients originate from HTLV-1 endemic areas, the West Indies and West Africa. Histological alterations and inflammation in patients muscles were mostly moderate, with classical features of idiopathic myositis and rare HTLV-1-infected infiltrating cells. In all patients, HTLV-1 belonged to the A subtype, transcontinental subgroup. Anti-HTLV-1 antibodies titers were high, but the proviral load was not elevated compared to asymptomatic HTLV-1 carriers. CONCLUSION: We show here that muscle inflammation is moderate in HAIM, and accompanied by a low HTLV-1 proviral load, suggesting that the pathogenetic events do not exactly mirror those of other HTLV-1-associated inflammatory diseases.


Subject(s)
HTLV-I Infections/virology , Human T-lymphotropic virus 1/isolation & purification , Inflammation/virology , Myositis/virology , Adult , Africa, Western , Aged , Aged, 80 and over , Female , Human T-lymphotropic virus 1/classification , Human T-lymphotropic virus 1/genetics , Humans , Male , Middle Aged , Phylogeny , Proviruses/isolation & purification , RNA, Messenger/analysis , RNA, Viral/analysis , Retrospective Studies , Statistics, Nonparametric , Viral Load , West Indies
20.
Blood ; 120(3): 572-80, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22589473

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) is the causative agent of adult T-cell leukemia/lymphoma and HTLV-1-associated myelopathy/tropical spastic paraparesis. In addition to blood transfusion and sexual transmission, HTLV-1 is transmitted mainly through prolonged breastfeeding, and such infection represents a major risk for the development of adult T-cell leukemia/lymphoma. Although HTLV-1-infected lymphocytes can be retrieved from maternal milk, the mechanisms of HTLV-1 transmission through the digestive tract remain unknown. In the present study, we assessed HTLV-1 transport across the epithelial barrier using an in vitro model. Our results show that the integrity of the epithelial barrier was maintained during coculture with HTLV-1-infected lymphocytes, because neither morphological nor functional alterations of the cell monolayer were observed. Enterocytes were not susceptible to HTLV-1 infection, but free infectious HTLV-1 virions could cross the epithelial barrier via a transcytosis mechanism. Such virions were able to infect productively human dendritic cells located beneath the epithelial barrier. Our data indicate that HTLV-1 crosses the tight epithelial barrier without disruption or infection of the epithelium to further infect target cells such as dendritic cells. The present study provides the first data pertaining to the mode of HTLV-1 transport across a tight epithelial barrier, as can occur during mother-to-child HTLV-1 transmission during breastfeeding.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/virology , HTLV-I Infections/metabolism , Human T-lymphotropic virus 1/metabolism , Transcytosis/physiology , Virion/metabolism , Caco-2 Cells , Coculture Techniques , Dendritic Cells/metabolism , Enterocytes/cytology , Enterocytes/metabolism , Enterocytes/virology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/virology , HEK293 Cells , HT29 Cells , HTLV-I Infections/transmission , HTLV-I Infections/virology , Humans , Microscopy, Electron, Transmission , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Tight Junctions/metabolism , Tight Junctions/ultrastructure , Tight Junctions/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...