Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Front Immunol ; 13: 842468, 2022.
Article in English | MEDLINE | ID: mdl-36248831

ABSTRACT

The role of the mucosal pulmonary antibody response in coronavirus disease 2019 (COVID-19) outcome remains unclear. Here, we found that in bronchoalveolar lavage (BAL) samples from 48 patients with severe COVID-19-infected with the ancestral Wuhan virus, mucosal IgG and IgA specific for S1, receptor-binding domain (RBD), S2, and nucleocapsid protein (NP) emerged in BAL containing viruses early in infection and persist after virus elimination, with more IgA than IgG for all antigens tested. Furthermore, spike-IgA and spike-IgG immune complexes were detected in BAL, especially when the lung virus has been cleared. BAL IgG and IgA recognized the four main RBD variants. BAL neutralizing titers were higher early in COVID-19 when virus replicates in the lung than later in infection after viral clearance. Patients with fatal COVID-19, in contrast to survivors, developed higher levels of mucosal spike-specific IgA than IgG but lost neutralizing activities over time and had reduced IL-1ß in the lung. Altogether, mucosal spike and NP-specific IgG and S1-specific IgA persisting after lung severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) clearance and low pulmonary IL-1ß correlate with COVID-19 fatal outcome. Thus, mucosal SARS-CoV-2-specific antibodies may have adverse functions in addition to protective neutralization. Highlights: Mucosal pulmonary antibody response in COVID-19 outcome remains unclear. We show that in severe COVID-19 patients, mucosal pulmonary non-neutralizing SARS-CoV-2 IgA persit after viral clearance in the lung. Furthermore, low lung IL-1ß correlate with fatal COVID-19. Altogether, mucosal IgA may exert harmful functions beside protective neutralization.


Subject(s)
COVID-19 , Interleukin-1beta/metabolism , SARS-CoV-2 , Antibodies, Viral , Antigen-Antibody Complex , Cross-Sectional Studies , Humans , Immunoglobulin A , Immunoglobulin G , Lung , Nucleocapsid Proteins , Spike Glycoprotein, Coronavirus
2.
iScience ; 24(12): 103478, 2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34841222

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) rapidly rampaged worldwide, causing a pandemic of coronavirus disease (COVID -19), but the biology of SARS-CoV-2 remains under investigation. We demonstrate that both SARS-CoV-2 spike protein and human coronavirus 229E (hCoV-229E) or its purified S protein, one of the main viruses responsible for the common cold, induce the transient opening of Pannexin-1 (Panx-1) channels in human lung epithelial cells. However, the Panx-1 channel opening induced by SARS-CoV-2 is greater and more prolonged than hCoV-229E/S protein, resulting in an enhanced ATP, PGE2, and IL-1ß release. Analysis of lung lavages and tissues indicate that Panx-1 mRNA expression is associated with increased ATP, PGE2, and IL-1ß levels. Panx-1 channel opening induced by SARS-CoV-2 spike protein is angiotensin-converting enzyme 2 (ACE-2), endocytosis, and furin dependent. Overall, we demonstrated that Panx-1 channel is a critical contributor to SARS-CoV-2 infection and should be considered as an alternative therapy.

3.
Front Immunol ; 12: 735922, 2021.
Article in English | MEDLINE | ID: mdl-34671353

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a major public health issue. COVID-19 is considered an airway/multi-systemic disease, and demise has been associated with an uncontrolled immune response and a cytokine storm in response to the virus. However, the lung pathology, immune response, and tissue damage associated with COVID-19 demise are poorly described and understood due to safety concerns. Using post-mortem lung tissues from uninfected and COVID-19 deadly cases as well as an unbiased combined analysis of histology, multi-viral and host markers staining, correlative microscopy, confocal, and image analysis, we identified three distinct phenotypes of COVID-19-induced lung damage. First, a COVID-19-induced hemorrhage characterized by minimal immune infiltration and large thrombus; Second, a COVID-19-induced immune infiltration with excessive immune cell infiltration but no hemorrhagic events. The third phenotype correspond to the combination of the two previous ones. We observed the loss of alveolar wall integrity, detachment of lung tissue pieces, fibroblast proliferation, and extensive fibrosis in all three phenotypes. Although lung tissues studied were from lethal COVID-19, a strong immune response was observed in all cases analyzed with significant B cell and poor T cell infiltrations, suggesting an exhausted or compromised immune cellular response in these patients. Overall, our data show that SARS-CoV-2-induced lung damage is highly heterogeneous. These individual differences need to be considered to understand the acute and long-term COVID-19 consequences.


Subject(s)
COVID-19/mortality , COVID-19/pathology , Lung Injury/pathology , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/pathology , Aged , Aged, 80 and over , Autopsy , CD8-Positive T-Lymphocytes/immunology , Cytokine Release Syndrome/mortality , Cytokine Release Syndrome/pathology , Epithelial Cells/pathology , Female , Hemorrhage/pathology , Humans , Inflammation/pathology , Lung/pathology , Lung Injury/virology , Lymphopenia/pathology , Macrophage Activation/immunology , Macrophages/immunology , Male , Middle Aged , Myocytes, Smooth Muscle/pathology , Neutrophils/immunology , SARS-CoV-2 , Thrombosis/pathology
4.
Blood ; 137(1): 89-102, 2021 01 07.
Article in English | MEDLINE | ID: mdl-32818241

ABSTRACT

The role of ribosome biogenesis in erythroid development is supported by the recognition of erythroid defects in ribosomopathies in both Diamond-Blackfan anemia and 5q- syndrome. Whether ribosome biogenesis exerts a regulatory function on normal erythroid development is still unknown. In the present study, a detailed characterization of ribosome biogenesis dynamics during human and murine erythropoiesis showed that ribosome biogenesis is abruptly interrupted by the decline in ribosomal DNA transcription and the collapse of ribosomal protein neosynthesis. Its premature arrest by the RNA Pol I inhibitor CX-5461 targeted the proliferation of immature erythroblasts. p53 was activated spontaneously or in response to CX-5461, concomitant to ribosome biogenesis arrest, and drove a transcriptional program in which genes involved in cell cycle-arrested, negative regulation of apoptosis, and DNA damage response were upregulated. RNA Pol I transcriptional stress resulted in nucleolar disruption and activation of the ATR-CHK1-p53 pathway. Our results imply that the timing of ribosome biogenesis extinction and p53 activation is crucial for erythroid development. In ribosomopathies in which ribosome availability is altered by unbalanced production of ribosomal proteins, the threshold downregulation of ribosome biogenesis could be prematurely reached and, together with pathological p53 activation, prevents a normal expansion of erythroid progenitors.


Subject(s)
Cell Differentiation/physiology , Erythroid Cells/cytology , Erythropoiesis/physiology , Ribosomes/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Hematopoietic Stem Cells , Humans , Mice , Organelle Biogenesis
5.
Sci Transl Med ; 12(535)2020 03 18.
Article in English | MEDLINE | ID: mdl-32188724

ABSTRACT

In addition to hemostasis, human platelets have several immune functions and interact with infectious pathogens including HIV in vitro. Here, we report that platelets from HIV-infected individuals on combined antiretroviral drug therapy (ART) with low blood CD4+ T cell counts (<350 cells/µl) contained replication-competent HIV despite viral suppression. In vitro, human platelets harboring HIV propagated the virus to macrophages, a process that could be prevented with the biologic abciximab, an anti-integrin αIIb/ß3 Fab. Furthermore, in our cohort, 88% of HIV-infected individuals on ART with viral suppression and with platelets containing HIV were poor immunological responders with CD4+ T cell counts remaining below <350 cells/µl for more than one year. Our study suggests that platelets may be transient carriers of HIV and may provide an alternative pathway for HIV dissemination in HIV-infected individuals on ART with viral suppression and poor CD4+ T cell recovery.


Subject(s)
Blood Platelets , HIV Infections , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes , HIV Infections/drug therapy , Humans , Macrophages , Viral Load
6.
Exp Hematol ; 63: 22-27.e3, 2018 07.
Article in English | MEDLINE | ID: mdl-29654952

ABSTRACT

Animal evidence that platelet production occurs in the lungs is growing. We have investigated whether there is evidence to support pulmonary platelet production from studies using human conditions. We documented the presence of megakaryocytes (MKs) in the human pulmonary circulation and analyzed the role of the vascular microenvironment on MK function. Our results suggest that the endothelial microenvironment favors platelet formation and that von Willebrand factor combined with appropriate physical forces in flowing blood are determinant for platelet release. We also demonstrate that MKs have the potential to change ploidy as they circulate. These findings demonstrate a new pathophysiological environment affecting platelet production and provide new targets for therapeutic intervention.


Subject(s)
Cellular Microenvironment/physiology , Endothelium, Vascular/cytology , Megakaryocytes/metabolism , Pulmonary Circulation/physiology , Thrombopoiesis/physiology , Cell Separation , Hemorheology , Human Umbilical Vein Endothelial Cells , Humans , Megakaryocytes/cytology , Platelet Glycoprotein GPIb-IX Complex , Polyploidy , Pulmonary Artery , von Willebrand Factor/physiology
7.
Shock ; 43(1): 74-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25255379

ABSTRACT

Sepsis induces alterations of coagulation suggesting both hypercoagulable or hypocoagulable features. The result of their combination remains unknown, making it difficult to predict whether one prevails over the other. Thrombin generation tests (TGTs) stand as an interesting tool to establish an integrative phenotype of coagulation. It has been reported that septic patients display a hypocoagulable trait using TGT. However, protein C (PC) system response was not evaluated. We aimed at describing the thrombin generation profile in patients with septic shock under conditions that are sensitive to PC system to evaluate the net results of coagulation abnormalities and to determine whether hypercoagulable or hypocoagulable traits coexist within a given individual. Thrombin generation was studied in plasma from patients presenting with septic shock at diagnosis and 6 h after a conventional therapeutic management using calibrated automated thrombography with or without thrombomodulin (TM) addition. Patients exhibit clear alterations of TGT that present as both consumption-related hypocoagulability (evidenced without TM addition) but also hypercoagulability by decreased sensitivity to the PC system evidenced with TM addition. No difference could be demonstrated between survivors and nonsurvivors at Day 28, but patients who do not respond to therapeutics at 6 h seem to be more hypercoagulable. More importantly, if our results evidence heterogeneity between patients, we show that alterations of coagulation result in an equilibrium in the majority of patients, thus suggesting "normocoagulability"; but, in the presence of a biological imbalance between baseline thrombin generation and sensitivity to TM, the global effect mostly tends toward hypercoagulability. Thus, TGT may help identify distinct biological coagulation phenotypes in the complex alterations induced by sepsis.


Subject(s)
Blood Coagulation , Protein C/metabolism , Shock, Septic , Aged , Disease-Free Survival , Female , Humans , Male , Middle Aged , Shock, Septic/blood , Shock, Septic/mortality , Shock, Septic/therapy , Survival Rate , Thrombin Time/instrumentation , Thrombin Time/methods , Thrombomodulin/metabolism , Time Factors
8.
Semin Thromb Hemost ; 37(6): 664-72, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22102269

ABSTRACT

Thrombocytopenia (TP) Cargeeg is a unique autosomal dominant disorder, affecting a seven-generation family, caused by cytochrome C (CYCS) mutation that dysregulates platelet formation. The CYCS mutation in this disorder is a glycine 41 replacement by serine, which yields a cytochrome C variant with enhanced apoptotic pathway activity in vitro. The deregulated apoptosis in this disorder affects megakaryocytes (MK) during platelet formation, leading to early and ectopic platelet release in the bone marrow (BM). Notably, the family has no other phenotypic indication of abnormal apoptosis, implying that cytochrome C activity is not a critical regulator of physiological apoptosis in most cells. The pathophysiology of this unique inherited TP, with unaltered platelet survival and normal MK content in the BM, has implications for physiological and pathological mechanisms altering MK apoptosis, with implications for other unexplained thrombocytopenic disorders.


Subject(s)
Apoptosis/genetics , Cytochromes c/genetics , Mutation, Missense , Thrombocytopenia/genetics , Base Sequence , Female , Humans , Male , Megakaryocytes/metabolism , Megakaryocytes/pathology , Pedigree , Thrombocytopenia/congenital , Thrombocytopenia/physiopathology
9.
Exp Hematol ; 39(6): 629-42, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21420467

ABSTRACT

OBJECTIVE: LYL-1 is a transcription factor containing a basic helix-loop-helix motif closely related to SCL/TAL-1, a regulator of erythroid differentiation. Because LYL-1 is expressed in erythroid cell populations, we addressed its role in erythropoiesis using knockin mice. MATERIALS AND METHODS: Erythropoiesis of LYL-1(-/-) mice was studied by progenitor assays, flow cytometry, reconstitution assays, and functional tests. Expression of LYL-1, SCL, and GATA-1 was assessed at messenger RNA level by quantitative reverse transcription polymerase chain reaction. RESULTS: LYL-1(-/-) mice displayed decreased erythropoiesis with a partial arrest in differentiation, and enhanced apoptosis associated with decreased Bcl-x(L) expression in the bone marrow (BM). In addition, LYL-1(-/-) BM cells were severely impaired in their abilities to reconstitute the erythroid lineage in competitive assays, suggesting a cell autonomous abnormality of erythropoiesis. In parallel, erythroid progenitor and precursor cells were significantly increased in the spleen of LYL-1(-/-) mice. Expression of LYL-1 was differentially regulated during maturation of erythroblasts and strikingly different between spleen- and BM-derived erythroblasts. Expression of LYL-1 decreased during erythroid differentiation in the spleen whereas it increased in the BM to reach the same level in mature erythroblasts as in the soleen. Loss of Lyl-1 expression was accompanied with an increase of SCL/TAL-1 and GATA-1 transcripts in spleen but not in BM-derived erythroblasts. Furthermore, phenylhydrazine-induced stress erythropoiesis was elevated in LYL-1(-/-) mice and mutant BM and spleen erythroid progenitors were hypersensitive to erythropoietin. CONCLUSIONS: Taken together, these results suggest that LYL-1 plays a definite role in erythropoiesis, albeit with different effects in BM specifically regulating basal erythropoiesis, and spleen, controlling stress-induced erythropoiesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Erythropoiesis/genetics , Neoplasm Proteins/physiology , Stress, Physiological , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , DNA Primers , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
10.
Blood ; 116(8): 1244-53, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20489054

ABSTRACT

Transforming growth factor-beta1 (TGF-beta1) is a pleiotropic cytokine with major in vitro effects on hematopoietic stem cells (HSCs) and lymphocyte development. Little is known about hematopoiesis from mice with constitutive TGF-beta1 inactivation largely because of important embryonic lethality and development of a lethal inflammatory disorder in TGF-beta1(-/-) pups, making these studies difficult. Here, we show that no sign of the inflammatory disorder was detectable in 8- to 10-day-old TGF-beta1(-/-) neonates as judged by both the number of T-activated and T-regulator cells in secondary lymphoid organs and the level of inflammatory cytokines in sera. After T-cell depletion, the inflammatory disease was not transplantable in recipient mice. Bone marrow cells from 8- to 10-day-old TGF-beta1(-/-) neonates showed strikingly impaired short- and long-term reconstitutive activity associated with a parallel decreased in vivo homing capacity of lineage negative (Lin(-)) cells. In addition an in vitro-reduced survival of immature progenitors (Lin(-) Kit(+) Sca(+)) was observed. Similar defects were found in liver cells from TGF-beta1(-/-) embryos on day 14 after vaginal plug. These data indicate that TGF-beta1 is a critical regulator for in vivo homeostasis of the HSCs, especially for their homing potential.


Subject(s)
Autoimmune Diseases/immunology , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , Inflammation/immunology , Transforming Growth Factor beta1/physiology , Animals , Animals, Newborn , Autoimmune Diseases/pathology , Blotting, Western , Bone Marrow Cells/pathology , Cell Lineage , Cell Separation , Cells, Cultured , Cytokines/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Fetus , Flow Cytometry , Inflammation/pathology , Male , Mice , Mice, Knockout , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
11.
Cell Mol Life Sci ; 67(4): 545-56, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20012669

ABSTRACT

Thrombocytopenia is a frequent complication of viral infections providing evidence that interaction of platelets with viruses is an important pathophysiological phenomenon. Multiple mechanisms are involved depending on the nature of the viruses involved. These include immunological platelet destruction, inappropriate platelet activation and consumption, and impaired megakaryopoiesis. Viruses bind platelets through specific receptors and identified ligands, which lead to mutual alterations of both the platelet host and the viral aggressor. We have shown that HIV-1 viruses are internalized specifically in platelets and megakaryocytes, where they can be either sheltered, unaltered (with potential transfer of the viruses into target organs), or come in contact with platelet secretory products leading to virus destruction and facilitated platelet clearance. In this issue, we have reviewed the various pathways that platelets use in order to interact with viruses, HIV and others. This review also shows that more work is still needed to precisely identify platelet roles in viral infections, and to answer the challenge of viral safety in platelet transfusion.


Subject(s)
Blood Platelets/immunology , Thrombocytopenia/virology , Virus Diseases/immunology , Viruses/immunology , Blood Platelets/virology , Cell Adhesion Molecules/immunology , HIV/immunology , HIV Infections/complications , HIV Infections/immunology , Humans , Lectins, C-Type/immunology , Megakaryocytes/immunology , Megakaryocytes/virology , Platelet Transfusion/adverse effects , Receptors, Cell Surface/immunology , Thrombocytopenia/immunology , Virus Diseases/complications
12.
Blood ; 114(9): 1875-83, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19525480

ABSTRACT

Platelets originate from megakaryocytes (MKs) by cytoplasmic elongation into proplatelets. Direct platelet release is not seen in bone marrow hematopoietic islands. It was suggested that proplatelet fragmentation into platelets can occur intravascularly, yet evidence of its dependence on hydrodynamic forces is missing. Therefore, we investigated whether platelet production from MKs could be up-regulated by circulatory forces. Human mature MKs were perfused at a high shear rate on von Willebrand factor. Cells were observed in real time by videomicroscopy, and by confocal and electron microscopy after fixation. Dramatic cellular modifications followed exposure to high shear rates: 30% to 45% adherent MKs were converted into proplatelets and released platelets within 20 minutes, contrary to static conditions that required several hours, often without platelet release. Tubulin was present in elongated proplatelets and platelets, thus ruling out membrane tethers. By using inhibitors, we demonstrated the fundamental roles of microtubule assembly and MK receptor GPIb. Secretory granules were present along the proplatelet shafts and in shed platelets, as shown by P-selectin labeling. Platelets generated in vitro were functional since they responded to thrombin by P-selectin expression and cytoskeletal reorganization. In conclusion, MK exposure to high shear rates promotes platelet production via GPIb, depending on microtubule assembly and elongation.


Subject(s)
Blood Platelets/cytology , Blood Platelets/metabolism , Megakaryocytes/cytology , Megakaryocytes/metabolism , Cell Membrane/metabolism , Cells, Cultured , Flow Cytometry/methods , Humans , Microscopy, Confocal/methods , Microscopy, Electron/methods , Microscopy, Video/methods , Models, Biological , Perfusion , Stress, Mechanical , Thrombopoiesis/immunology , Tubulin/metabolism , von Willebrand Factor/metabolism
13.
Semin Thromb Hemost ; 35(2): 213-23, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19408194

ABSTRACT

The electron microscope has proved to be a useful tool to study and understand the biology of platelets and to classify many platelet disorders. After a technical overview, this article reviews syndromes originating from platelet organelle, cytoskeleton, and membrane defects for which electron microscopy plays a role in the diagnostic process, such as gray platelet syndrome, Paris-Trousseau syndrome, storage pool diseases, MYH9-related thrombocytopenias, or Wiskott-Aldrich syndrome. Particular focus is given to the ultrastructural aspect of platelets in these disorders.


Subject(s)
Blood Platelet Disorders/blood , Blood Platelet Disorders/diagnosis , Microscopy, Electron, Transmission/methods , Blood Platelets/metabolism , Blood Platelets/ultrastructure , Humans
14.
Blood ; 113(7): 1535-42, 2009 Feb 12.
Article in English | MEDLINE | ID: mdl-19029443

ABSTRACT

Quebec platelet disorder (QPD) is an inherited bleeding disorder associated with increased urokinase plasminogen activator (uPA) in platelets but not in plasma, intraplatelet plasmin generation, and alpha-granule protein degradation. These abnormalities led us to investigate uPA expression by QPD CD34(+) progenitors, cultured megakaryocytes, and platelets, and whether uPA was stored in QPD alpha-granules. Although QPD CD34(+) progenitors expressed normal amounts of uPA, their differentiation into megakaryocytes abnormally increased expression of the uPA gene but not the flanking genes for vinculin or calcium/calmodulin-dependent protein kinase IIgamma on chromosome 10. The increased uPA production by cultured QPD megakaryocytes mirrored their production of alpha-granule proteins, which was normal. uPA was localized to QPD alpha-granules and it showed extensive colocalization with alpha-granule proteins in both cultured QPD megakaryocytes and platelets, and with plasminogen in QPD platelets. In QPD megakaryocytes, cultured without or with plasma as a source of plasminogen, alpha-granule proteins were stored undegraded and this was associated with much less uPA-plasminogen colocalization than in QPD platelets. Our studies indicate that the overexpression of uPA in QPD emerges with megakaryocyte differentiation, without altering the expression of flanking genes, and that uPA is costored with alpha-granule proteins prior to their proteolysis in QPD.


Subject(s)
Blood Platelet Disorders/pathology , Blood Platelet Disorders/physiopathology , Megakaryocyte Progenitor Cells/cytology , Megakaryocyte Progenitor Cells/physiology , Urokinase-Type Plasminogen Activator/genetics , Antigens, CD34/metabolism , Blood Platelets/cytology , Blood Platelets/physiology , Cell Differentiation/physiology , Cells, Cultured , Gene Expression/physiology , Humans , Plasminogen/metabolism , Thrombopoiesis/physiology , Urokinase-Type Plasminogen Activator/metabolism
15.
Nat Genet ; 40(4): 387-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18345000

ABSTRACT

We report the first identified mutation in the gene encoding human cytochrome c (CYCS). Glycine 41, invariant throughout eukaryotes, is substituted by serine in a family with autosomal dominant thrombocytopenia caused by dysregulated platelet formation. The mutation yields a cytochrome c variant with enhanced apoptotic activity in vitro. Notably, the family has no other phenotypic indication of abnormal apoptosis, implying that cytochrome c activity is not a critical regulator of most physiological apoptosis.


Subject(s)
Apoptosis/physiology , Cytochromes c/genetics , Mutation/genetics , Signal Transduction , Thrombocytopenia/etiology , Apoptotic Protease-Activating Factor 1/metabolism , Female , Genetic Linkage , Humans , Male , Megakaryocytes/metabolism , Megakaryocytes/pathology , Oxidation-Reduction , Pedigree , Platelet Count , Serine/chemistry , Serine/genetics , Thrombocytopenia/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...