Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters










Publication year range
1.
Viruses ; 16(1)2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38257782

ABSTRACT

Coagulation disorders are described in COVID-19 and long COVID patients. In particular, SARS-CoV-2 infection in megakaryocytes, which are precursors of platelets involved in thrombotic events in COVID-19, long COVID and, in rare cases, in vaccinated individuals, requires further investigation, particularly with the emergence of new SARS-CoV-2 variants. CD147, involved in the regulation of inflammation and required to fight virus infection, can facilitate SARS-CoV-2 entry into megakaryocytes. MCT4, a co-binding protein of CD147 and a key player in the glycolytic metabolism, could also play a role in SARS-CoV-2 infection. Here, we investigated the susceptibility of megakaryocytes to SARS-CoV-2 infection via CD147 and MCT4. We performed infection of Dami cells and human CD34+ hematopoietic progenitor cells induced to megakaryocytic differentiation with SARS-CoV-2 pseudovirus in the presence of AC-73 and syrosingopine, respective inhibitors of CD147 and MCT4 and inducers of autophagy, a process essential in megakaryocyte differentiation. Both AC-73 and syrosingopine enhance autophagy during differentiation but only AC-73 enhances megakaryocytic maturation. Importantly, we found that AC-73 or syrosingopine significantly inhibits SARS-CoV-2 infection of megakaryocytes. Altogether, our data indicate AC-73 and syrosingopine as inhibitors of SARS-CoV-2 infection via CD147 and MCT4 that can be used to prevent SARS-CoV-2 binding and entry into megakaryocytes, which are precursors of platelets involved in COVID-19-associated coagulopathy.


Subject(s)
Megakaryocytes , Phenols , Reserpine , SARS-CoV-2 , Humans , COVID-19 , Megakaryocytes/virology , Phenols/pharmacology , Post-Acute COVID-19 Syndrome , Reserpine/analogs & derivatives , Reserpine/pharmacology , SARS-CoV-2/drug effects , Virus Internalization/drug effects
2.
Front Cell Infect Microbiol ; 11: 715208, 2021.
Article in English | MEDLINE | ID: mdl-34513730

ABSTRACT

Dengue virus (DENV) infection can cause either self-limited dengue fever or hemorrhagic complications. Low platelet count is one of the manifestations of dengue fever. Megakaryocytes are the sole producers of platelets. However, the role of both host and viral factors in megakaryocyte development, maturation, and platelet production is largely unknown in DENV infection. PI3K/AKT/mTOR pathway plays a significant role in cell survival, maturation, and megakaryocyte development. We were interested to check whether pathogenic insult can impact this pathway. We observed decreased expression of most of the major key molecules associated with the PI3K/AKT/mTOR pathway in DENV infected MEG-01 cells. In this study, the involvement of PI3K/AKT/mTOR pathway in megakaryocyte development and maturation was confirmed with the use of specific inhibitors in infected MEG-01 cells. Our results showed that direct pharmacologic inhibition of this pathway greatly impacted megakaryopoiesis associated molecule CD61 and some essential transcription factors (GATA-1, GATA-2, and NF-E2). Additionally, we observed apoptosis in megakaryocytes due to DENV infection. Our results may suggest that DENV impairs PI3K/AKT/mTOR axis and molecules involved in the development and maturation of megakaryocytes. It is imperative to investigate the role of these molecules in the context of megakaryopoiesis during DENV infection to better understand the pathways and mechanisms, which in turn might provide insights into the development of antiviral strategies.


Subject(s)
Dengue Virus , Megakaryocytes , Signal Transduction , Transcription Factors/metabolism , Cell Line , Humans , Megakaryocytes/metabolism , Megakaryocytes/virology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism
3.
Immunol Lett ; 236: 31-36, 2021 08.
Article in English | MEDLINE | ID: mdl-34111476

ABSTRACT

Activation of innate receptors in megakaryocytes (MKs) may affect the ability to produce functional platelets. Low platelet count is one of the clinical manifestations of dengue virus (DENV) infection. In MKs, the effect of innate receptors during DENV-infection is not well studied. Here we used MEG-01 cells to investigate DENV serotype 2 induced innate receptors in these cells. DENV RNA was estimated by qRT-PCR in the culture supernatant. The expression of innate receptors was determined by western blot and qPCR. DENV infection led to increased expression of RIG-I at 24 hrs post-infection (hpi) and MDA-5 at 48 and 72 hpi (p<0.05). However, no change in the expression of TLR3 at protein level was observed. Activation of MDA-5 resulted in increased expression of IFN-ß and ISG-15 in DENV infected MEG-01 cells, which was further confirmed by MDA-5 siRNA treatment. Apart from inducing innate receptors, DENV significantly decreases the expression of CD61, an activation marker of megakaryocyteson MEG-01 cells as observed by flow cytometry analysis (p<0.01). Results from this study confirm that DENV infection activates the type-I interferon in megakaryocytes and may play a significant role in maturation and development.


Subject(s)
Dengue Virus/physiology , Dengue/etiology , Dengue/metabolism , Disease Susceptibility , Host-Pathogen Interactions/immunology , Megakaryocytes/immunology , Megakaryocytes/metabolism , Animals , Biomarkers , Cell Line , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Immunity, Innate , Immunophenotyping , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/metabolism , Interferon-beta/biosynthesis , Megakaryocytes/virology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
4.
J Hematol Oncol ; 14(1): 72, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33926500

ABSTRACT

Evidence suggests that platelets may directly interact with SARS-CoV-2, raising the concern whether ACE2 receptor plays a role in this interaction. The current study showed that SARS-CoV-2 interacts with both platelets and megakaryocytes despite the limited efficiency. Abundance of the conventional receptor ACE2 and alternative receptors or co-factors for SARS-CoV-2 entry was characterized in platelets from COVID-19 patients and healthy persons as well as human megakaryocytes based on laboratory tests or previously reported RNA-seq data. The results suggest that SARS-CoV-2 interacts with platelets and megakaryocytes via ACE2-independent mechanism and may regulate alternative receptor expression associated with COVID-19 coagulation dysfunction.


Subject(s)
Blood Platelets/virology , COVID-19/blood , Megakaryocytes/virology , Receptors, Virus/physiology , SARS-CoV-2/physiology , Virus Attachment , Angiotensin-Converting Enzyme 2/analysis , Blood Platelets/metabolism , COVID-19/complications , COVID-19/genetics , Cell Line , Humans , Megakaryocytes/metabolism , Organ Specificity , Platelet Activation , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Viral/isolation & purification , SARS-CoV-2/isolation & purification , Thrombophilia/etiology , Thrombophilia/virology , Transcription, Genetic , Virus Internalization
5.
J Cell Physiol ; 236(9): 6726-6741, 2021 09.
Article in English | MEDLINE | ID: mdl-33611789

ABSTRACT

Thrombocytopenia is a common complication of human cytomegalovirus (HCMV) infection in immunocompromised hosts, which contributes to poor prognosis even in patients receiving antiviral treatment. Here, we investigated the megakaryo/thrombopoiesis process, including the involvement of the c-Mpl/IEX-1 pathway, after HCMV infection, identified receptors mediating the interaction between megakaryocytes (MKs) and HCMV, and explored novel therapeutic targets. Our data shows that HCMV directly infects megakaryocytes in patients with HCMV DNAemia and influences megakaryopoiesis via the c-Mpl/IEX-1 pathway throughout megakaryocyte maturation, apoptosis, and platelet generation in vivo and in vitro. After treatment with inhibitors of PDGFRα and αvß3, the HCMV infection rate in MKs was significantly reduced, suggesting that IMC-3G3 and anti-αvß3 are potential therapeutic alternatives for viral infection. In summary, our study proposes a possible mechanism and potential treatments for thrombocytopenia caused by HCMV infection and other viral diseases associated with abnormal hemostasis.


Subject(s)
Cytomegalovirus/physiology , Hematopoietic Stem Cell Transplantation , Integrin alphaVbeta3/metabolism , Megakaryocytes/virology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptors, Thrombopoietin/metabolism , Signal Transduction , Thrombopoiesis , Adolescent , Adult , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Child , Cytomegalovirus/ultrastructure , Cytomegalovirus Infections/pathology , Down-Regulation , Female , Humans , Male , Membrane Proteins/metabolism , Middle Aged , Multivariate Analysis , Ploidies , Risk Factors , Toll-Like Receptor 2/metabolism , Transplantation, Homologous , Young Adult
6.
Virchows Arch ; 478(3): 487-496, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32915265

ABSTRACT

Pulmonary megakaryocytes participate in the pathogenesis of lung damage, particularly in acute lung injury. Although megakaryocytes are not mentioned as a characteristic histologic finding associated to pulmonary injury, a few studies reveal that their number is increased in diffuse alveolar damage (DAD). In this autopsy study, we have observed a relevant number of pulmonary megakaryocytes in COVID-19 patients dying with acute lung injury (7.61 ± 5.59 megakaryocytes per 25 high-power fields vs. 1.14 ± 0.86 for the control group, p < 0.05). We analyzed samples of 18 patients, most of whom died after prolonged disease and use of mechanical ventilation. Most patients showed advanced DAD and abnormal coagulation parameters with high levels of fibrinogen, D-dimers, and variable thrombocytopenia. For comparison, pulmonary samples from a group of 14 non-COVID-19 patients dying with DAD were reviewed. They showed similar pulmonary histopathologic findings and an increase in the number of megakaryocytes (4 ± 4.17 vs. 1.14 ± 0.86 for the control group, p < 0.05). Megakaryocyte count in the COVID-19 group was greater but did not reach statistical significance (7.61 ± 5.59 vs. 4 ± 4.17, p = 0.063). Regardless of the cause, pulmonary megakaryocytes are increased in patients with DAD. Their high number seen in COVID-19 patients suggests a relation with the thrombotic events so often seen these patients. Since the lung is considered an active site of megakaryopoiesis, a prothrombotic status leading to platelet activation, aggregation and consumption may trigger a compensatory pulmonary response.


Subject(s)
COVID-19/pathology , SARS-CoV-2/physiology , Thrombosis/pathology , Adult , Aged , Autopsy , COVID-19/virology , Female , Humans , Lung/pathology , Lung/virology , Male , Megakaryocytes/pathology , Megakaryocytes/virology , Middle Aged , Thrombosis/virology
7.
J Thromb Haemost ; 19(1): 46-50, 2021 01.
Article in English | MEDLINE | ID: mdl-33119197

ABSTRACT

There is an urgent need to understand the underlying mechanisms contributing to thrombotic and inflammatory complications during COVID-19. Data from independent groups have identified that platelets are hyperreactive during COVID-19. Platelet hyperreactivity is accompanied by changes in platelet gene expression, and enhanced interactions between platelets and leukocytes. In some patients, SARS-CoV-2 mRNA has been detected in platelets. Together, this suggests that SARS-CoV-2 may interact with platelets. However, controversy remains on which receptors mediate SARS-CoV-2 platelet interactions. Most, but not all, transcriptomic and proteomic analyses fail to observe the putative SARS-CoV-2 receptor, angiotensin converting enzyme-2, or the cellular serine protease necessary for viral entry, TMPRSS2, on platelets and megakaryocytes. Interestingly, platelets express other known SARS-CoV-2 receptors, which induce similar patterns of activation to those observed when platelets are incubated with SARS-CoV-2. This article explores these findings and discusses ongoing areas of controversy and uncertainty with regard to SARS-CoV-2 platelet interactions.


Subject(s)
Angiotensin-Converting Enzyme 2/blood , Blood Platelets/virology , COVID-19/blood , COVID-19/virology , Receptors, Virus/blood , SARS-CoV-2/physiology , SARS-CoV-2/pathogenicity , Angiotensin-Converting Enzyme 2/physiology , COVID-19/complications , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Humans , Megakaryocytes/virology , Models, Biological , Platelet Activation , RNA, Viral/blood , RNA, Viral/genetics , Receptors, Virus/physiology , SARS-CoV-2/genetics , Serine Endopeptidases/blood , Serine Endopeptidases/physiology , Thrombosis/blood , Thrombosis/etiology , Thrombosis/virology , Virus Internalization
8.
Sci Rep ; 10(1): 19587, 2020 11 11.
Article in English | MEDLINE | ID: mdl-33177556

ABSTRACT

Dengue virus (DENV) infection causes dengue fever in humans, which can lead to thrombocytopenia showing a marked reduction in platelet counts, and dengue hemorrhagic fever. The virus may cause thrombocytopenia either by destroying the platelets or by interfering with their generation via the process of megakaryopoiesis. MEG-01 is the human megakaryoblastic leukemia cell line that can be differentiated in vitro by phorbol-12-myristate-13-acetate (PMA) treatment to produce platelet-like-particles (PLPs). We have studied DENV infection of MEG-01 cells to understand its effect on megakaryopoiesis and the generation of PLPs. We observed that DENV could infect only naive MEG-01 cells, and differentiated cells were refractory to virus infection/replication. However, DENV-infected MEG-01 cells, when induced for differentiation with PMA, supported an enhanced viral replication. Following the virus infection, the MEG-01 cells showed a marked reduction in the surface expression of platelet markers (CD41, CD42a, and CD61), a decreased polyploidy, and significantly reduced PLP counts. DENV infection caused an enhanced Notch signaling in MEG-01 cells where the virus envelope protein was shown to interact with TAL-1, a host protein important for megakaryopoiesis. These observations provide new insight into the role of DENV in modulating the megakaryopoiesis and platelet production process.


Subject(s)
Dengue/blood , Host-Pathogen Interactions/physiology , T-Cell Acute Lymphocytic Leukemia Protein 1/metabolism , Thrombopoiesis/physiology , Viral Envelope Proteins/metabolism , Blood Platelets/physiology , Blood Platelets/virology , Cell Differentiation/drug effects , Cell Line, Tumor , Dengue/virology , Dengue Virus/pathogenicity , Humans , Leukemia, Megakaryoblastic, Acute/pathology , Megakaryocytes/virology , Polyploidy , Receptors, Notch/metabolism , Tetradecanoylphorbol Acetate/pharmacology
9.
Platelets ; 31(8): 1085-1089, 2020 Nov 16.
Article in English | MEDLINE | ID: mdl-32857624

ABSTRACT

Coronavirus disease 2019 (COVID-19) is a global public health emergency with many clinical facets, and new knowledge about its pathogenetic mechanisms is deemed necessary; among these, there are certainly coagulation disorders. In the history of medicine, autopsies and tissue sampling have played a fundamental role in order to understand the pathogenesis of emerging diseases, including infectious ones; compared to the past, histopathology can be now expanded by innovative techniques and modern technologies. For the first time in worldwide literature, we provide a detailed postmortem and biopsy report on the marked increase, up to 1 order of magnitude, of naked megakaryocyte nuclei in the bone marrow and lungs from serious COVID-19 patients. Most likely related to high interleukin-6 serum levels stimulating megakaryocytopoiesis, this phenomenon concurs to explain well the pulmonary abnormal immunothrombosis in these critically ill patients, all without molecular or electron microscopy signs of megakaryocyte infection.


Subject(s)
Betacoronavirus/pathogenicity , Bone Marrow/pathology , Coronavirus Infections/pathology , Cytokine Release Syndrome/pathology , Disseminated Intravascular Coagulation/pathology , Lung/pathology , Pneumonia, Viral/pathology , Thrombosis/pathology , Adult , Aged , Autopsy , Betacoronavirus/immunology , Bone Marrow/immunology , Bone Marrow/virology , COVID-19 , Cell Nucleus/immunology , Cell Nucleus/pathology , Cell Nucleus/virology , Coronavirus Infections/complications , Coronavirus Infections/immunology , Coronavirus Infections/virology , Critical Illness , Cytokine Release Syndrome/complications , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/virology , Disseminated Intravascular Coagulation/complications , Disseminated Intravascular Coagulation/immunology , Disseminated Intravascular Coagulation/virology , Fatal Outcome , Host-Pathogen Interactions/immunology , Humans , Interleukin-6/biosynthesis , Interleukin-6/immunology , Lung/immunology , Lung/virology , Male , Megakaryocytes/immunology , Megakaryocytes/pathology , Megakaryocytes/virology , Middle Aged , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , SARS-CoV-2 , Severity of Illness Index , Thrombopoiesis/immunology , Thrombosis/complications , Thrombosis/immunology , Thrombosis/virology
10.
Med Hypotheses ; 143: 110098, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32688185

ABSTRACT

The neoteric severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been jeopardizing the world with the symptoms of seasonal flu. The virus contagion predicted to have been originated from Wuhan, China has by far trapped 4,198,418 cases from 212 countries in the world with two international conveyances with 284,102 deaths as of 11 May 2020 (10:18 GMT). Researchers around the globe have indulged in deciphering viral mode in the body for devising a cure. Affirmations from autopsies and preliminary findings on SARS-CoV-2 hypothesized on viral pathogenesis within the host, for instance, source of inflammation in lungs and pneumonia. This hypothesis assigns the platelets as agents of infection after viral entry. Presently, curbing infection to stall the spread of SARS-CoV-2 is the prima facie intervention employed, worldwide. However, public health authorities must monitor the state of affairs scrupulously, as the deeper our understanding of this novel virus and its associated outbreak, the better we can deal with it. Knowing this idea might be far-fetched, yet this postulate would serve as the groundwork for the present situation.


Subject(s)
Blood Platelets/virology , Coronavirus Infections/blood , Lung/virology , Pneumonia, Viral/blood , Betacoronavirus , COVID-19 , China , Cytokines/metabolism , Humans , Inflammation/blood , Inflammation/virology , Megakaryocytes/virology , Pandemics , SARS-CoV-2
11.
J Virol ; 94(6)2020 02 28.
Article in English | MEDLINE | ID: mdl-31852793

ABSTRACT

Human herpesvirus 6B (HHV-6B), a T-lymphotropic virus, infects almost exclusively humans. An animal model of HHV-6B has not been available. Here, we report the first animal model to mimic HHV-6B pathogenesis; the model is based on humanized mice in which human immune cells were engrafted and maintained. For HHV-6B replication, adequate human T-cell activation (which becomes susceptible to HHV-6B) is necessary in this murine model. Here, we found that an additional transfer of human mononuclear cells to humanized mice resulted in an explosive proliferation of human activated T cells, which could be representative of graft-versus-host disease (GVHD) because the primary transfer of human cells was not sufficient to increase the number and ratio of human T cells. Mice infected with HHV-6B became weak and/or died approximately 7 to 14 days later. Quantitative PCR analysis revealed that the spleen and lungs were the major sites of HHV-6B replication in this model, and this was corroborated by the detection of viral proteins in these organs. Histological analysis also revealed the presence of megakaryocytes, indicating HHV-6B infection. Multiplex analysis of cytokines/chemokines in sera from the infected mice showed secretions of human cytokines/chemokines as reported for both in vitro infection and clinical samples, indicating that the secreted cytokines could affect pathogenesis. This is the first animal model showing HHV-6B pathogenesis, and it will be useful for elucidating the pathogenicity of HHV-6B, which is related to GVHD and idiopathic pneumonia syndrome.IMPORTANCE Human herpesvirus 6B (HHV-6B) is a ubiquitous virus that establishes lifelong latent infection only in humans, and the infection can reactivate, with severe complications that cause major problems. A small-animal model of HHV-6B infection has thus been desired for research regarding the pathogenicity of HHV-6B and the development of antiviral agents. We generated humanized mice by transplantation with human hematopoietic stem cells, and here, we modified the model by providing an additional transfer of human mononuclear cells, providing the proper conditions for efficient HHV-6B infection. This is the first humanized mouse model to mimic HHV-6B pathogenesis, and it has great potential for research into the in vivo pathogenesis of HHV-6B.


Subject(s)
Graft vs Host Disease/immunology , Herpesvirus 6, Human/immunology , Pneumonia, Viral/immunology , Roseolovirus Infections/immunology , Animals , Cell Line , Disease Models, Animal , Graft vs Host Disease/pathology , Graft vs Host Disease/virology , Humans , Megakaryocytes/immunology , Megakaryocytes/pathology , Megakaryocytes/virology , Mice , Mice, Knockout , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Roseolovirus Infections/pathology , Syndrome , T-Lymphocytes/immunology , T-Lymphocytes/pathology , T-Lymphocytes/virology
12.
PLoS Negl Trop Dis ; 13(11): e0007837, 2019 11.
Article in English | MEDLINE | ID: mdl-31765380

ABSTRACT

One of the most important clinical signs of dengue virus infection is the reduction of white blood cells and platelets in human peripheral blood (leukopenia and thrombocytopenia, respectively), which may significantly impair the clearance of dengue virus by the immune system. The cause of thrombocytopenia and leukopenia during dengue infection is still unknown, but may be related to severe suppression of bone marrow populations including hematopoietic stem cells and megakaryocytes, the progenitors of white blood cells and platelets respectively. Here, we explored the possibility that bone marrow suppression, including ablation of megakaryocyte populations, is caused by dengue virus infection of megakaryocytes. We used three different models to measure dengue virus infection and replication: in vitro, in a human megakaryocyte cell line with viral receptors, ex vivo, in primary human megakaryocytes, and in vivo, in humanized mice. All three systems support dengue virus infection and replication, including virus strains from serotypes 1, 2, and 3, and clinical signs, in vivo; all assays showed viral RNA and/or infectious viruses 7-14 days post-infection. Although we saw no significant decrease in cell viability in vitro, there was significant depletion of mature megakaryocytes in vivo. We conclude that megakaryocytes can produce dengue viruses in the bone marrow niche, and a reduction of cell numbers may affect bone marrow homeostasis.


Subject(s)
Dengue Virus/growth & development , Dengue/pathology , Megakaryocytes/virology , Animals , Bone Marrow/virology , Cells, Cultured , Disease Models, Animal , Female , Humans , Male , Mice , Mice, SCID , Models, Theoretical
13.
Transfusion ; 59(9): 2938-2951, 2019 09.
Article in English | MEDLINE | ID: mdl-31251408

ABSTRACT

BACKGROUND: Dengue virus (DENV) is a significant threat to public health in tropical and subtropical regions, where the frequency of human migration is increasing. Transmission of DENV from donors to recipients after hematopoietic stem cell transplantation has been steadily described. However, the underlying mechanisms remain unclear. STUDY DESIGN AND METHODS: Freshly isolated bone marrow (BM) was subjected to DENV infection, followed by multicolor fluorescence-activated cell sorting (FACS) analysis. Virus in supernatants was collected and analyzed by plaque assay. RESULTS: DENV-1 to DENV-4 could effectively infect freshly obtained BM and produced infectious virus. DENV infection did not change the quantitative population of hematopoietic stem and progenitor cells (HSPCs), megakaryocytic progenitor cells (MkPs) and megakaryocytes. Additionally, DENV antigen, nonstructural protein 1, was enriched in HSPCs and MkPs of DENV infected marrow cells. CD34+, CD133+, or CD61+ cells sorted out from BM were not only the major contributing targets facilitating the DENV infection directly but also facilitated the spread of DENV into other cells when cocultured. CONCLUSION: Results suggest that DENV can efficiently infect HSPCs, which might jeopardize the recipients if DENV-infected cells were subsequently used. We therefore raise the need for DENV screening for both the donors and recipients of hematopoietic stem cell transplantation, especially for donors exposed to endemic areas, to mitigate DENV infection in immunocompromised recipients.


Subject(s)
Dengue Virus/growth & development , Dengue/pathology , Dengue/transmission , Hematopoietic Stem Cells/virology , Viral Plaque Assay , Antigens, Viral/analysis , Antigens, Viral/isolation & purification , Bone Marrow Cells/pathology , Bone Marrow Cells/physiology , Bone Marrow Cells/virology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Dengue/blood , Dengue Virus/pathogenicity , Fetal Blood/cytology , Fetal Blood/virology , Flow Cytometry , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/physiology , Humans , Immunophenotyping , Megakaryocytes/pathology , Megakaryocytes/physiology , Megakaryocytes/virology , Myeloid Progenitor Cells/pathology , Myeloid Progenitor Cells/physiology , Myeloid Progenitor Cells/virology
14.
Virol J ; 15(1): 115, 2018 07 28.
Article in English | MEDLINE | ID: mdl-30055639

ABSTRACT

BACKGROUND: Bovine viral diarrhea virus (BVDV) causes significant economic losses worldwide in the cattle industry through decrease in productive performance and immunosuppression of animals in herds. Recent studies conducted by our group showed that mice can be infected with BVDV-1 by the oral route. The purpose of this study was to assess the clinical signs, hematological changes, histopathological lesions in lymphoid tissues, and the distribution of the viral antigen after oral inoculation with a Korean noncytopathic (ncp) BVDV-2 field isolate in mice. METHODS: Mice were orally administered a low or high dose of BVDV-2; blood and tissue samples were collected on days 2, 5, and 9 postinfection (pi). We monitored clinical signs, hematological changes, histopathological lesions, and tissue distribution of a viral antigen by reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry (IHC) and then compared these parameters with those in ncp BVDV-1 infections. RESULTS: None of the infected mice developed any clinical signs of the illness. Significant thrombocytopenia was found in both low- and high-dose-inoculated mice on day 2 pi. Leukopenia was apparent only in low-dose-inoculated mice on day 2 pi, whereas lymphopenia was not observed in any ncp BVDV-2-infected animal. Viral RNA was found in the spleen in of low- and high-dose-inoculated mice by RT-PCR. According to the results of IHC, the viral antigen was consistently detected in lymphocytes of bone marrow and spleen and less frequently in bronchus-associated lymphoid tissue (BALT), mesenteric lymph nodes, and Peyer's patches. Despite the antigen detection in BALT and mesenteric lymph nodes, histopathological lesions were not observed in these tissues. Lympholysis, infiltration by inflammatory cells, and increased numbers of megakaryocytes were seen in Peyer's patches, spleens, and bone marrow, respectively. In contrast to ncp BVDV-1 infection, lympholysis was found in the spleen of ncp BVDV-2-infected mice. These histopathological lesions were more severe in high-dose-inoculated mice than in low-dose-inoculated mice. CONCLUSIONS: Our results provide insight into the pathogenesis of ncp BVDV-2 infection in mice. Collectively, these results highlight significant differences in pathogenesis between ncp BVDV-1 and ncp BVDV-2 infections in a murine model.


Subject(s)
Bone Marrow/pathology , Diarrhea Virus 2, Bovine Viral/physiology , Megakaryocytes/pathology , Megakaryocytes/virology , Pestivirus Infections/pathology , Pestivirus Infections/virology , Animals , Cattle , Disease Models, Animal , Hemorrhagic Syndrome, Bovine/blood , Hemorrhagic Syndrome, Bovine/pathology , Hemorrhagic Syndrome, Bovine/virology , Mice , Pestivirus Infections/blood , Peyer's Patches/pathology , Peyer's Patches/virology , RNA, Viral , Spleen/pathology , Spleen/virology , Viral Load
15.
Sci Rep ; 5: 17990, 2015 Dec 11.
Article in English | MEDLINE | ID: mdl-26657027

ABSTRACT

The levels of neutralizing antibody to a pathogen are an effective indicator to predict efficacy of a vaccine in trial. And yet not all the trial vaccines are in line with the theory. Using dengue virus (DENV) to investigate the viral morphology affecting the predictive value, we evaluated the viral morphology in acute dengue plasma compared to that of Vero cells derived DENV. The virions in plasma were infectious and heterogeneous in shape with a "sunny-side up egg" appearance, viral RNA was enclosed with CD61+ cell-derived membrane interspersed by the viral envelope protein, defined as dengue vesicles. The unique viral features were also observed from ex vivo infected human bone marrow. Dengue vesicles were less efficiently neutralized by convalescent patient serum, compared to virions produced from Vero cells. Our results exhibit a reason why potencies of protective immunity fail in vivo and significantly impact dengue vaccine and drug development.


Subject(s)
Dengue Virus/physiology , Dengue/metabolism , Dengue/virology , Integrin beta3/metabolism , Animals , Biological Transport , Bone Marrow Cells/metabolism , Bone Marrow Cells/virology , Capsid Proteins/metabolism , Cell Membrane/metabolism , Chlorocebus aethiops , Cytoplasmic Vesicles/metabolism , Cytoplasmic Vesicles/virology , Dengue/immunology , Dengue Virus/classification , Dengue Virus/isolation & purification , Dengue Virus/ultrastructure , Humans , Megakaryocytes/metabolism , Megakaryocytes/virology , Phenotype , Serogroup , Vero Cells , Viral Load , Virion/ultrastructure
16.
J Virol ; 87(21): 11648-58, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23966397

ABSTRACT

A characteristic clinical feature of dengue virus infection is thrombocytopenia, though its underlying mechanism is not definitively determined. By adoptive transfer of human CD34(+) fetal liver cells into immunodeficient mice, we have constructed humanized mice with significant levels of human platelets, monocytes/macrophages, and hepatocytes. Infection of these mice with both lab-adapted and clinical strains of dengue virus induces characteristic human hematological changes, including transient leukopenia and thrombocytopenia. We show that the specific depletion of human platelets is not mediated by antibodies in the periphery or reduced production of human thrombopoietin in the liver but reduction of human megakaryocytes and megakaryocyte progenitors in the bone marrow of the infected mice. These findings identify inhibition of platelet production in the bone marrow as a key mechanism underlying dengue-induced thrombocytopenia and suggest the utility of the improved humanized mouse model in studying dengue virus infection and pathogenesis in a human cell context.


Subject(s)
Bone Marrow/pathology , Bone Marrow/virology , Dengue Virus/pathogenicity , Dengue/complications , Megakaryocytes/physiology , Megakaryocytes/virology , Thrombocytopenia/etiology , Animals , Humans , Mice , Mice, SCID
17.
Int J Hematol ; 96(5): 600-10, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22987294

ABSTRACT

Although hematological disorders with salient features of thrombocytopenia have been well documented in dengue patients, the role of CD61-expressing platelets and the megakaryocytic cell lineage in the pathogenesis of dengue virus (DENV) infection remains largely unexplored. A prospective observational study was performed using blood samples and PBMCs from dengue-confirmed patients, as well as from rhesus monkeys (RM) experimentally infected with DENV. Immunohistochemical staining and FACS techniques were applied to evaluate the frequencies of CD61(+) cells that contained DENV antigen. Highly enriched population of CD61(+) cells was also isolated from acute DENV-infected RM and assayed for DENV RNA by quantitative RT-PCR. Results revealed that DENV antigen was found in small vesicles of varying size, and more frequently in anucleated cells associated with platelets in dengue patients. The DENV antigen-containing cells were CD61(+) and appeared to share characteristics of megakaryocytes. Kinetic profiles of CD61(+) cells from DENV-infected RM revealed a transient increase in CD61(+)CD62P(+) cells early after DENV infection. DENV RNA in a highly enriched population of CD61(+) cells from the infected RM was observed during acute stage. Our results indicate that virus containing CD61(+) cells may be directly linked to the platelet dysfunction and low platelet count characteristics of dengue patients.


Subject(s)
Blood Platelets , Dengue Virus/metabolism , Dengue , Integrin beta3/blood , Megakaryocytes , Thrombocytopenia , Acute Disease , Animals , Antigens, Viral/blood , Blood Platelets/metabolism , Blood Platelets/virology , Dengue/blood , Dengue/complications , Female , Humans , Macaca mulatta , Male , Megakaryocytes/metabolism , Megakaryocytes/virology , Prospective Studies , RNA, Viral/blood , Thrombocytopenia/blood , Thrombocytopenia/etiology , Thrombocytopenia/virology
18.
Exp Hematol ; 40(3): 250-259.e4, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22193689

ABSTRACT

Abnormal bone marrow (BM) suppression is one of the hallmarks of dengue virus (DENV) infection in patients. Although the etiology remains unclear, direct viral targeting of the BM has been reasoned to be a contributing factor. The present studies were carried out in an effort to determine the potential effect of DENV infection on the cellularity of BM using a previously established nonhuman primate model of DENV-induced coagulopathy. BM aspirates were collected at various times from the infected nonhuman primate and cells were phenotypically defined and isolated using standard flow cytometry (fluorescence-activated cell sorting). These isolated cells were subjected to detection of DENV utilizing quantitative real-time reverse transcription polymerase chain reaction, electron microscopy, and immunostaining techniques. DENV RNA was detectable by quantitative real-time reverse transcription polymerase chain reaction in BM specimens and the presence of DENV-like particles within platelet was confirmed by electron microscopy. Enumeration of BM cells revealed a transient surge in cellularity at day 1, followed by a gradual decline from days 2 to 10 post infection. Detailed phenotypic studies showed similar kinetics in the frequencies of CD41(+)CD61(+) cells, regardless of CD34 and CD45 expression. The CD61(+) cells were not only the predominant cells that stained for DENV antigen but fluorescence-activated cell sorting-assisted isolation of CD61(+) cells from the BM were shown to contain infectious DENV by coculture with Vero cells. These data support the view that intravenous infection of nonhuman primate with DENV leads to direct infection of the BM, which is likely to be a contributing factor for transient cell suppression in the peripheral blood characteristic of acute DENV infection.


Subject(s)
Bone Marrow Cells/virology , Dengue Virus/physiology , Dengue/virology , Animals , Antigens, CD/analysis , Blood Platelets/ultrastructure , Blood Platelets/virology , Bone Marrow Cells/ultrastructure , Cell Lineage , Chlorocebus aethiops , Coculture Techniques , Dengue/blood , Dengue/pathology , Dengue Virus/ultrastructure , Giant Cells/virology , Immunophenotyping , Macaca mulatta , Megakaryocytes/virology , Microscopy, Electron , Plasma/virology , RNA, Viral/analysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Vero Cells/virology , Viral Load , Viremia/virology
19.
PLoS One ; 7(12): e52902, 2012.
Article in English | MEDLINE | ID: mdl-23300812

ABSTRACT

Depression of the peripheral blood platelet count during acute infection is a hallmark of dengue. This thrombocytopenia has been attributed, in part, to an insufficient level of platelet production by megakaryocytes that reside in the bone marrow (BM). Interestingly, it was observed that dengue patients experience BM suppression at the onset of fever. However, few studies focus on the interaction between dengue virus (DENV) and megakaryocytes and how this interaction can lead to a reduction in platelets. In the studies reported herein, BM cells from normal healthy rhesus monkeys (RM) and humans were utilized to identify the cell lineage(s) that were capable of supporting virus infection and replication. A number of techniques were employed in efforts to address this issue. These included the use of viral RNA quantification, nonstructural protein and infectivity assays, phenotypic studies utilizing immunohistochemical staining, anti-differentiation DEAB treatment, and electron microscopy. Cumulative results from these studies revealed that cells in the BM were indeed highly permissive for DENV infection, with human BM having higher levels of viral production compared to RM. DENV-like particles were predominantly observed in multi-nucleated cells that expressed CD61+. These data suggest that megakaryocytes are likely the predominant cell type infected by DENV in BM, which provides one explanation for the thrombocytopenia and the dysfunctional platelets characteristic of dengue virus infection.


Subject(s)
Bone Marrow Cells/virology , Bone Marrow/virology , Cell Lineage/physiology , Dengue/virology , Integrin beta3/metabolism , Animals , Bone Marrow/metabolism , Bone Marrow Cells/metabolism , Colony-Forming Units Assay , Dengue/metabolism , Dengue Virus , Humans , Macaca mulatta , Megakaryocytes/metabolism , Megakaryocytes/virology , Thrombocytopenia/metabolism , Thrombocytopenia/virology , p-Aminoazobenzene/analogs & derivatives , p-Aminoazobenzene/pharmacology
20.
Virology ; 405(1): 70-80, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20673746

ABSTRACT

Hantaan virus (HTNV), the prototype member of the Hantavirus genus in the family Bunyaviridae, causes hemorrhagic fever with renal syndrome (HFRS) in humans. Hemorrhage is due to endothelial barrier damage and a sharp decrease in platelet counts. The mechanisms underlying HTNV-associated acute thrombocytopenia have not been elucidated so far. Platelets are produced by mature megakaryocytes that develop during megakaryopoiesis. In this study, we show that HTNV targets megakaryocytic cells whereas rather non-pathogenic hantaviruses did not infect this cell type. After induction of differentiation megakaryocytic cells switched from low-level to high-level HTNV production without reduction in cell survival or alteration in differentiation. However, increased HTNV replication resulted in strong upregulation of HLA class I molecules although HTNV escaped type I interferon (IFN)-associated innate responses. Taken together, HTNV efficiently replicates in differentiating megakaryocytic cells resulting in upregulation of HLA class I molecules, the target structures for cytotoxic T cells (CTLs).


Subject(s)
Gene Expression Regulation, Viral/physiology , Genes, MHC Class I/physiology , Hantaan virus/pathogenicity , Megakaryocytes/cytology , Megakaryocytes/virology , Virus Replication/physiology , Animals , Cell Adhesion , Cell Proliferation , Chlorocebus aethiops , Genes, MHC Class I/genetics , Humans , Up-Regulation , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...