Figure 3 - uploaded by Juan Carlos Zapata
Content may be subject to copyright.
Platelet content. Membrane glycoproteins of platelets include GPIa, GPIIb/IIIa (aIIbb3), or VLA-5 (fibrinogen receptor); GPIb/IX/V (vW and Mac-1 receptor); GPIc’-IIa or VLA-6 (laminin receptor); and a2b1 GPVI (Collagen receptor). Alpha granules contain P-selectin; platelet factor 4; transforming factor-b1; chemokines; proteoglycan; platelet-derived growth factor; a2-plasmin inhibitor; vitronectin; laminin; CD63; TGFbeta; CLEC-2; thrombospondin; fibronectin; B-thromboglobulin; vWF; fibrinogen; coagulation factors V, XI, and XIII; integrins; thrombocidins; proteases; thrombin; prothrombin; kininogens; immunoglobulin family receptors; leucine-rich repeat family receptors; and other proinflammatory and immune- modulating factors. Dense granules hold ADP, ATP, calcium, serotonin, histamine, dopamine, phosphate, eicosanoids. Receptors for primary agonist include P2X, P2Y 1 , and P2Y 12 (ADP); TPa-R and TPb-R (TXA 2 ); PAR-1 and PAR-4 (thrombin); PAFR (platelet-activating factor); 5-HT 2A 

Platelet content. Membrane glycoproteins of platelets include GPIa, GPIIb/IIIa (aIIbb3), or VLA-5 (fibrinogen receptor); GPIb/IX/V (vW and Mac-1 receptor); GPIc’-IIa or VLA-6 (laminin receptor); and a2b1 GPVI (Collagen receptor). Alpha granules contain P-selectin; platelet factor 4; transforming factor-b1; chemokines; proteoglycan; platelet-derived growth factor; a2-plasmin inhibitor; vitronectin; laminin; CD63; TGFbeta; CLEC-2; thrombospondin; fibronectin; B-thromboglobulin; vWF; fibrinogen; coagulation factors V, XI, and XIII; integrins; thrombocidins; proteases; thrombin; prothrombin; kininogens; immunoglobulin family receptors; leucine-rich repeat family receptors; and other proinflammatory and immune- modulating factors. Dense granules hold ADP, ATP, calcium, serotonin, histamine, dopamine, phosphate, eicosanoids. Receptors for primary agonist include P2X, P2Y 1 , and P2Y 12 (ADP); TPa-R and TPb-R (TXA 2 ); PAR-1 and PAR-4 (thrombin); PAFR (platelet-activating factor); 5-HT 2A 

Source publication
Article
Full-text available
Viral hemorrhagic fevers (VHF) are acute zoonotic diseases that, early on, seem to cause platelet destruction or dysfunction. Here we present the four major ways viruses affect platelet development and function and new evidence of molecular factors that are preferentially induced by the more pathogenic members of the families Flaviviridae, Bunyavir...

Context in source publication

Context 1
... recently, West Africa), with very high case fatality rates, ranging from 25% to 90% [38,39]. These viruses are most often transmitted by direct contact with infected animals or people. The natural reservoirs for filoviruses are thought to be bats [38,40]. Only two vaccines are licensed to prevent VHF: YF17D and Candid # 1, live-attenuated versions of the YF virus and JUNV. Unfortunately, the VHF-endemic areas are expanding, with the aggravation that treatments are limited or nonexistent. For this reason, it is important to continue research and development for new prevention and control options. VHF signs and symptoms range from asymptomatic infection to life-threatening disease. Initially, patients develop a nonspecific febrile syndrome (flu-like) that can include chills, malaise, headache, backache, arthralgia, myalgia, retro-orbital pain, anorexia, nausea, vomiting, diarrhea, cough, and sore throat [41–46]. At this stage the clinical signs resemble other infectious diseases, making early VHF clinical diagnoses impossible. Some cases progress to severe disease characterized by systemic vascular damage that, depending on the virus, will manifest as subcutaneous bleeding (flushing, conjunctivitis, peri-orbital edema, petechiae, or ecchymosis), positive tourni- quet test, hypotension and internal organ bleeding, hematem- esis, and melena [41–46]. The cause of bleeding in most VHF diseases is a disseminated intravascular coagulation (DIC) that depletes the coagulation factors, inducing massive plasma leakage, hypovolemia, and shock. A prolonged shock state leads to multiple organ failures and, in some cases, death. However, the loss of blood is rarely the cause of death (with the exception of filoviruses) [41–46]. The hallmark characteristic of all VHF is a decrease in platelet numbers and/or function. This decrease is usually accompanied by an increase in fibrinogen degradation products, prothrombin time (PT), and partial thromboplastin time (PTT). At least five days after the onset of fever, there is also a marked leucopenia and high viral loads that are associated with fatal outcomes. Another common finding is an increase in the alanine aminotransferase (ALT) and aspartate aminotransferase (AST) enzymes, primarily due to liver damage [41–46]. Based on some clinical and laboratory findings (Table 3), different pathogenic mechanisms have been proposed for each VHF, including depletion of hepatic coagulation factors, cytokine storm, increased permeability by vascular endothelial growth factor, complement activation, and DIC. In spite of the differences seen with each VHF, there is a large body of evidence indicating that viral replication and host immune responses play an important role in determining disease severity and clinical outcome [47–50]. HF viruses can establish nonlytic replication, or ‘‘virus factories,’’ in monocytes and/or macrophage and DC [47,49]. Viral replication subverts the function of these cells, as well as the function of uninfected bystander cells, to undermine the innate immune response, such as interferon (IFN) production. This leads to uncontrolled viral replication and to a lack of specific antiviral responses in the host [47–50]. These cells might also act as vehicles to carry the virus to its replication sites, such as endothelium, liver, spleen, and other organs, leading to the pantropism of most hemorrhagic fever viruses [47–50]. Whereas the early replication stage is characterized by a lack of IFN production and IFN-responsive gene expression, later viremic stages reveal an unregulated release of pro-inflammatory cytokines, such as tumor necrosis factor alpha (TNF- a ), IL-10, IL-1R a , and soluble TNF-R, that could contribute to immunosuppression and increased vascular permeability, leading to hemorrhagic signs [51,52]. Thrombocytopenia (reduction of platelet numbers) and depressed immune responses are hallmarks of VHF, and platelets are involved in both processes; therefore, this review will focus on the platelet’s role in VHF pathogenesis. Platelets, also known as thrombocytes, are small, 2–3 m m, enucleated cell fragments derived from the cytoplasm of large, 100 m m, megakaryocytes (MKs) located in the bone marrow (Figure 2) [53]. Each MK sheds thousands of platelets into the blood stream, making them the second-most-abundant cell type in peripheral blood. Platelets’ most-known function is to maintain the integrity of the vascular system [53]; however, their function as immune elements is becoming more evident. Under specific stimulus, such as tissue damage, platelets are activated by extracellular matrix proteins, such as collagen and von Willebrand factor. Their irregular shapes swell to a compact spherical form with projections containing glycoprotein receptors that help them to attach to each other and to cells at wound sites [54]. After activation, platelets release granule contents and coagulation factors that further enhance their activation and cell attachment (Figure 3). This process results in the formation of an effective plug at the site of injury that supports formation of a fibrin network and stops bleeding [55]. There are some pathological processes in which the normal number of circulating platelets can decrease (thrombocytopenia), increase (thrombocytosis), or diminish in function (thrombasthe- nia) with or without hemostatic manifestations. The study of those conditions showed that in addition to their role in homeostasis, platelets are important mediators of inflammation and innate immunity [56]. Sub-products generated from microorganisms, from complement fixation reactions, or from expression of receptors such as Fc and C3 a /C5 a can attract platelets to the site of infection or injury where they are activated by thrombin and bind to vascular endothelial cells and leukocytes. Platelets can also bind pathogens directly, recognizing them through Toll-like receptors (TLRs), and can alert other cells via the innate immune response [57]. Platelets can directly release antimicrobial factors such as platelet factor 4 (PF-4), RANTES, connective tissue activating peptide 3 (CTAP-3), platelet basic protein, thymosin b -4 (T b -4), fibrinopeptide B (FP-B), fibrinopeptide A (FP-A), superox- ide, hydrogen peroxide, and hydroxyl free radicals. Additionally, platelets participate in antibody-dependent cell cytotoxicity against microbial pathogens [58]. In conclusion, any pathogenic state affecting platelets will not only impact hemostasis but also will modify the immune response to infection. Although there are some advances in understanding the specific platelet activation mechanisms induced by some microorganisms, little is known about these interactions and their role in the pathogenesis of VHF. Several microorganisms or molecules derived from them interact with platelets and affect their function [55,59,60]. Virus–platelet interactions were initially described around 50 years ago with some in vitro experiments using influenza virus and other myxoviruses [61,62]. Since then, other viruses have been reported to interact not only with platelets but also with MKs [63– 68]. From these interactions, platelet numbers or functions can be compromised, and little is known about the exact mechanisms. In the case of VHF, recent publications described pathogenic mechanisms involving the role of platelets in homeostasis as well as their role in the initial immune responses in animals and human beings. The next paragraphs will describe different ways in which hemorrhagic fever viruses affect platelets. There are four major causes of thrombocytopenia or platelet malfunction induced by viruses. These include direct effects of viruses on platelets, immunological platelet destruction, impaired megakaryopoiesis, and MK destruction (Table 4) [68]. In the VHF diseases, the most common mechanism of thrombocytopenia is platelet disappearance from damaged tissues or generalized virus- induced DIC, in which coagulation factors are depleted. This common pattern does not seem to apply to LASV. Additionally, there is some evidence that viruses not belonging to the hemorrhagic fever group (such as varicella, herpes zoster, smallpox, rubella, measles, cytomegalovirus, rotavirus, adenovirus, and HIV), under specific circumstances, can also cause hemostatic problems and can be associated with DIC [66,69–72]. More work is necessary to clarify the role of the platelet–virus interaction in coagulopathies and in DIC induced by both hemorrhagic and nonhemorrhagic viruses. Platelets bind viruses through different receptors, such as b -3 integrins or TLRs, and platelets are known to express TLR2, TLR4, and TLR9 [73,74]. In severe sepsis, there are coagulation abnormalities and DIC that are thought to be due to TLR signaling in platelets [57]. Bacterial stimulation of platelet TLR2 increased P-selectin surface expression, activation of integrin a IIb b 3 , generation of reactive oxygen species, and formation of platelet–neutrophil heterotypic aggregates in human whole ...

Citations

... • YFV can pass through blood-brain barrier [57] • Leukopenia and Neutropenia [45] • Elevated TNFalpha [45], IL6, RANTES [56] • Neutralizing antibodies are primary source of protection. Reinfection in humans has not been documented [45] • Mild Thrombocytopenia [58,59] • Loss of factors II, V, VII, IX, and X, fibrinogen [60,61] • dysregulation of the plasmin system [60] • Elevated TT and d-Dimer seen in all cases. Fatal yellow fever associated with elevated APTT, loss of PRC, loss of FII, V, VII, IX [62] (Continued.) ...
... • Viremia ranges from undetectable to 50% mosquito infectious dose (10 ∧ 3 copies), to 10 ∧ 8.5MID [64] depending on host health, serotype. • Interacts with HS, HSP90, CD14, GRP78 and LamR, and (on myeloid cells) DC-SIGN, mannose receptor, ICAM-3, CLEC5 [65] • Can infect many cell lines in vitro [66], but only replicates in skin, pbmcs, spleen, lymph nodes, liver, possibly CNS, lung, and heart [67] • Monocytes, macrophages, and lymphocytes are early sites of replication [50,51] • and vWF release [69] • Cytokine tsunami (high levels of IL2, IL4, IL6, IL8, IL13 and IL18, TNFα and IFNγ) increases capillary permeability [68] • DENV reactive antibodies can cross react and damage endothelia [68] • DENV infection of endothelial cells drives destruction of basement membrane by MMP3, MMP13, MMP9 and some MMP2 [51,54,70,71] • Neutropenia and lymphocytosis [64] low leukocytes but increased lymphocyte numbers [69] • Strain mismatched antibodies can promote infection of monocytes [64] • T cells are protective but strain mismatched T cells may [72] contribute to cytokine overload • Denv infected monocytes and mast cells promote vascular leakage [68,73] • described as a 'cytokine tsunami' [74,75] with high levels of TNFa and IL6 [76,77] causing capillary leak and pleural effusion [78][79][80] • MCP1 [49], VEGF and loss of plasma VEGFR2 [81,82] • IFN signaling key to blocking DENV replication [66] • Elevated kynurenine [83] • Thrombocytopenia [64] • DENV van activate plasminogen [84] • DENV reactive antibodies can cross react with thrombin preventing clotting and plasminogen resulting in greater fibrinolysis [85][86][87][88] • Platelet crosslinking to endothelia [58,89] • Loss of serum serotonin [83] • DENV infection inhibits PGI-2 and ET-1, increases TM, vWF, and tPA [68] • DENV infection in children: low C, S, antithrombin III, increased TM, TF, PAI-1, TM correlated with shock severity, PAI-1 with bleeding severity [85] • Variable reduction in prothrombin, V, VII, VIII, XI, X, antithrombin and a2 antiplasmin. ...
... The pathogenesis of HFVs is intimately associated with coagulation; these pathogens are defined by hemorrhagic manifestations of bleeding, bruising, and plasma volume loss. Loss of platelets is found in EBOV, YFV, DFV, Junin (JV), SNV, PUUV, CCHFV [26, 58,64,109,115,119,122,160,161,171,172], and frank disseminated intravascular coagulation [243] is seen in CCHFV and PUUV [160,161,170,172]. The exception among HFVs reviewed here is LFV [98,[104][105][106][107]. ...
Article
Full-text available
The hemorrhagic fever viruses (HFVs) cause severe or fatal infections in humans. Named after their common symptom hemorrhage, these viruses induce significant vascular dysfunction by affecting endothelial cells, altering immunity, and disrupting the clotting system. Despite advances in treatments, such as cytokine blocking therapies, disease modifying treatment for this class of pathogen remains elusive. Improved understanding of the pathogenesis of these infections could provide new avenues to treatment. While animal models and traditional 2D cell cultures have contributed insight into the mechanisms by which these pathogens affect the vasculature, these models fall short in replicating in vivo human vascular dynamics. The emergence of microphysiological systems (MPSs) offers promising avenues for modeling these complex interactions. These MPS or ‘organ-on-chip’ models present opportunities to better mimic human vascular responses and thus aid in treatment development. In this review, we explore the impact of HFV on the vasculature by causing endothelial dysfunction, blood clotting irregularities, and immune dysregulation. We highlight how existing MPS have elucidated features of HFV pathogenesis as well as discuss existing knowledge gaps and the challenges in modeling these interactions using MPS. Understanding the intricate mechanisms of vascular dysfunction caused by HFV is crucial in developing therapies not only for these infections, but also for other vasculotropic conditions like sepsis.
... Acute zoonotic diseases known as viral hemorrhagic fevers (VHFs) initially appear to be related to platelet malfunction or destruction [1]. When Soviet researchers were examining hantaviral hemorrhagic fever (HF) with renal syndrome in the 1930s, they developed the idea of virus hemorrhagic fevers (VHFs). ...
Article
Full-text available
The term "Viral Hemorrhagic Fever" (VHF) describes a severe feverish sickness characterized by aberrant vascular control, vascular damage, and hemorrhagic symptoms. Multiple viruses belonging to distinct families are the cause of this illness. The viruses that cause VHF are categorized into seven distinct families according to the International Committee on Taxonomy of Viruses' most recent classification: Hantaviridae, Nairoviridae, Filoviridae, Phenuiviridae, Paramyxoviridae, Arenavidae, and Flaviviridae are the families involved. The concept of virus hemorrhagic fevers (VHFs) originated in the 1930s when Soviet researchers were studying hantaviral hemorrhagic fever (HF) with renal dysfunction. Dengue fever/Dengue haemorrhagic fever and Kyasanur forest sickness are the two most common viral hemorrhagic fevers (VHF) in India that are transmitted by arthropod vectors. The diagnosis of community-acquired pneumonia (CCHF) in India is greatly hampered by the co-occurring symptoms of hemorrhagic fevers such as Review Article Srivastav et al..; Asian J. Res. Infect. Dis., vol. 15, no. 3, pp. 17-25, 2024; Article no.AJRID114632 18 dengue, Kyasanur forest sickness, Hantavirus hemorrhagic fever, and other illnesses such as leptospirosis, meningococcal infections, and malaria. The pathophysiology, aetiology, diagnosis, treatment, symptoms, and indicators of virus hemorrhagic fevers (VHFs) are all covered in this review article.
... Acute zoonotic diseases known as viral hemorrhagic fevers (VHFs) initially appear to be related to platelet malfunction or destruction [1]. When Soviet researchers were examining hantaviral hemorrhagic fever (HF) with renal syndrome in the 1930s, they developed the idea of virus hemorrhagic fevers (VHFs). ...
Article
Full-text available
The term "Viral Hemorrhagic Fever" (VHF) describes a severe feverish sickness characterized by aberrant vascular control, vascular damage, and hemorrhagic symptoms. Multiple viruses belonging to distinct families are the cause of this illness. The viruses that cause VHF are categorized into seven distinct families according to the International Committee on Taxonomy of Viruses' most recent classification: Hantaviridae, Nairoviridae, Filoviridae, Phenuiviridae, Paramyxoviridae, Arenavidae, and Flaviviridae are the families involved. The concept of virus hemorrhagic fevers (VHFs) originated in the 1930s when Soviet researchers were studying hantaviral hemorrhagic fever (HF) with renal dysfunction. Dengue fever/Dengue haemorrhagic fever and Kyasanur forest sickness are the two most common viral hemorrhagic fevers (VHF) in India that are transmitted by arthropod vectors. The diagnosis of community-acquired pneumonia (CCHF) in India is greatly hampered by the co-occurring symptoms of hemorrhagic fevers such as Review Article Srivastav et al..; Asian J. Res. Infect. Dis., vol. 15, no. 3, pp. 17-25, 2024; Article no.AJRID114632 18 dengue, Kyasanur forest sickness, Hantavirus hemorrhagic fever, and other illnesses such as leptospirosis, meningococcal infections, and malaria. The pathophysiology, aetiology, diagnosis, treatment, symptoms, and indicators of virus hemorrhagic fevers (VHFs) are all covered in this review article.
... VHFs are all accompanied by thrombocytopenia. VHF consists of the family of Filoviruses (Ebola and Marburg), Arenaviruses (Lassa and New World arenaviruses), Bunyaviruses (Congo-Crimean Hemorrhagic Fever and Rift Valley Fever), and Flaviviruses (yellow fever) [45]. The geographical distribution of VHFs is constrained by the habitats of their natural hosts, with human beings serving as incidental hosts. ...
... Outbreaks of VHF occur sporadically, leading to these viruses consistently emerging or re-emerging in locations where optimal conditions are present. Dengue virus, along with Yellow fever (YF) virus, another flavivirus, ranks as one of the most globally distributed arthropod-borne diseases, holding the first and second positions, respectively [45]. ...
... Additionally, at least five days after the onset of fever, a pronounced leucopenia and heightened viral loads are observed, often correlating with fatal outcomes. Another common occurrence is an increase in alanine aminotransferase (ALT) and aspartate aminotransferase (AST) enzymes, primarily indicative of liver damage [45]. The crucial aspects of platelet behavior in response to infections and their intricate involvement in the immune response has been declared. ...
Article
Full-text available
Thrombocytopenia, characterized by a decrease in platelet count, is a multifaceted clinical manifestation that can arise from various underlying causes. This review delves into the intriguing nexus between viruses and thrombocytopenia, shedding light on intricate pathophysiological mechanisms and highlighting the pivotal role of platelets in viral infections. The review further navigates the landscape of thrombocytopenia in relation to specific viruses, and sheds light on the diverse mechanisms through which hepatitis C virus (HCV), measles virus, parvovirus B19, and other viral agents contribute to platelet depletion. As we gain deeper insights into these interactions, we move closer to elucidating potential therapeutic avenues and preventive strategies for managing thrombocytopenia in the context of viral infections.
... Flaviviruses infections often result in a decrease in the platelet count as result of direct viruses toxicity, immunological platelet destruction, megakaryocytes (cells responsible for platelets production in the bone marrow) destruction and impaired megakaryopoiesis. 38 Early and transient drop in platelet levels following 17D/ 17DD vaccine has been reported in healthy adults and PLWH. 8,11 Finally, minimal liver laboratorial abnormalities were found in this study. ...
Article
Full-text available
Background Safety data on the yellow fever vaccine 17DD in People Living with HIV (PLWH) are limited. This study explored the occurrence of post-vaccination 17DD viremia and the kinetics of hematological and liver laboratorial parameters in PLWH and HIV-uninfected participants [HIV(-) controls]. Methods We conducted a secondary analysis of a longitudinal interventional trial (NCT03132311) study that enrolled PLWH and HIV(-) controls to receive a single 17DD dose and were followed at 5, 30 and 365 days after vaccination in Rio de Janeiro, Brazil. 17DD viremia (obtained throughreal-time PCR and plaque forming units’ assays), hematological (neutrophils, lymphocytes and platelets counts) and liver enzymes (ALT and AST) results were assessed at baseline and Days 5 and 30 post-vaccination. Logistic regression models explored factors associated with the odds of having positive 17DD viremia. Linear regression models explored variables associated with hematological and liver enzymes results at Day 5. Results A total of 202 PLWH with CD4 ≥ 200 cells/µL and 68 HIV(-) controls were included in the analyses. 17DD viremia was found in 20.0 % of the participants and was twice more frequent in PLWH than in HIV(-) controls (22.8% vs. 11.8 %, p-value < 0.001). Neutrophils, lymphocytes and platelets counts dropped at Day 5 and returned to baseline values at Day 30. 17DD viremia was associated with lower nadir of lymphocytes and platelets at Day 5. ALT levels did not increase post-vaccination and were not associated with 17DD viremia. Conclusions 17DD was safe and well-tolerated in PLWH with CD4 ≥ 200 cells/µL. Post-vaccination viremia was more frequent in PLWH than in controls. Transient and self-limited decreases in lymphocytes and neutrophils occurred early after vaccination. 17DD viremia was associated with lower lymphocytes and platelets nadir after vaccination. We did not observe elevations in ALT after 17DD vaccination.
... During infection with a virus, activated platelets adhere to the sub-endothelium, and their hyperactivity causes thrombus formation, which can trigger arterial ischemia and even pulmonary embolisms. Like many other viruses such as the influenza virus (H1N1), human cytomegalovirus (HCMV) (6), immunodeficiency virus (HIV) (7), dengue virus (8,9), hepatitis C virus (HCV) (10) and Ebola (11) and many other viruses (12), SARS-CoV-2 (13)(14)(15)(16) can also directly lead to platelet hyperactivity. Zhang et al. (2020) showed that platelets from COVID-19 patients express ACE2 and TMPRSS2, and the Spike protein of SARS-CoV-2 can bind to platelet ACE2 and increase platelet activation (13). ...
Article
Full-text available
Introduction Pro-thrombotic events are one of the prevalent causes of intensive care unit (ICU) admissions among COVID-19 patients, although the signaling events in the stimulated platelets are still unclear. Methods We conducted a comparative analysis of platelet transcriptome data from healthy donors, ICU, and non-ICU COVID-19 patients to elucidate these mechanisms. To surpass previous analyses, we constructed models of involved networks and control cascades by integrating a global human signaling network with transcriptome data. We investigated the control of platelet hyperactivation and the specific proteins involved. Results Our study revealed that control of the platelet network in ICU patients is significantly higher than in non-ICU patients. Non-ICU patients require control over fewer proteins for managing platelet hyperactivity compared to ICU patients. Identification of indispensable proteins highlighted key subnetworks, that are targetable for system control in COVID-19-related platelet hyperactivity. We scrutinized FDA-approved drugs targeting indispensable proteins and identified fostamatinib as a potent candidate for preventing thrombosis in COVID-19 patients. Discussion Our findings shed light on how SARS-CoV-2 efficiently affects host platelets by targeting indispensable and critical proteins involved in the control of platelet activity. We evaluated several drugs for specific control of platelet hyperactivity in ICU patients suffering from platelet hyperactivation. The focus of our approach is repurposing existing drugs for optimal control over the signaling network responsible for platelet hyperactivity in COVID-19 patients. Our study offers specific pharmacological recommendations, with drug prioritization tailored to the distinct network states observed in each patient condition. Interactive networks and detailed results can be accessed at https://fostamatinib.bioinfo-wuerz.eu/.
... Установлено также, что хантавирусы могут и прямо активировать нейтрофилы через β 2 -интегрины этих клеток [20]. Через β 2 -интегрины нейтрофилов в дальнейшем происходит также активация тромбоцитов [21], а следствием агрегации тромбоцитов на поверхности нейтрофилов служит все тот же NET-оз, а также тромбоцитопения, провоцирующая геморрагические проявления болезни [22]. Моноциты/макрофаги служат своеобразным мостом между врожденным и адаптивным иммунитетом в связи с их важнейшей функцией презентации антигенов [23]. ...
Article
Full-text available
Hemorrhagic fever with renal syndrome (HFRS) is an acute natural focal disease of a viral nature. Its severity is determined by hemorrhagic phenomena, cytokine storm, kidney damage with the development of acute renal failure, and the possibility of death. HFRS is caused by viruses from the family Hantaviridae. Hantaviruses are characterized by the ability to infect endothelial and epithelial cells of the renal tubules, as well as innate immunity cells (neutrophils, monocytes, dendritic cells), which leads to both direct damage to the immune system and indirect through a violation of their functions. The review analyzes the immune mechanisms in HFRS and their role in the pathogenesis of this disease.
... Human infection with hemorrhagic fever disease-associated viruses (HFVs), such as Crimean-Congo hemorrhagic fever virus (CCHFV), Hantaan virus, Ebola virus, and Lassa virus, can lead to thrombocytopenia, resulting in severe bleeding in patients [18][19][20][21]. Thrombocytopenia induced by infection with the above mentioned HFVs was found to occur via four main mechanisms, including destruction of platelets by direct interaction with virus, immunological destruction of platelet-virus complexes, impairment of megakaryocytes and progenitor cells to produce platelets, and inhibition of platelet function [22]. However, the understanding of the mechanisms underlying platelet reduction caused by infection with HFVs is still insu cient [22]. ...
... Thrombocytopenia induced by infection with the above mentioned HFVs was found to occur via four main mechanisms, including destruction of platelets by direct interaction with virus, immunological destruction of platelet-virus complexes, impairment of megakaryocytes and progenitor cells to produce platelets, and inhibition of platelet function [22]. However, the understanding of the mechanisms underlying platelet reduction caused by infection with HFVs is still insu cient [22]. Infection with non-HFVs, such as SARS-CoV-2, can induce mild thrombocytopenia and thrombotic diathesis. ...
Preprint
Full-text available
Background Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne viral hemorrhagic fever disease caused by infection with Dabie bandavirus (SFTS virus, SFTSV). Thrombocytopenia is the primary clinical feature of SFTS and is significantly associated with disease severity. However, the pathological mechanism of thrombocytopenia in SFTS remains unclear. Methods Platelets purified from SFTS patients were subjected to RNA transcriptome analyses. Differentially expressed genes (DEGs) in the platelets of deceased and surviving patients were identified, and their functions and transcription levels were characterized. DEGs related to cell death were compared with the platelets of COVID-19 and dengue fever patients. The percentage of platelets positive for biomarkers of pyroptosis, apoptosis, necroptosis, autophagy, and ferroptosis was determined by flow cytometry. RNA transcriptome analyses were also performed with platelets purified from nonlethal SFTSV infection model mice. DEGs representing the functional changes in mouse platelets were characterized, and platelet death was also investigated. Functional platelet changes in SFTS patients and SFTSV-infected mice were compared to determine the different mechanisms underlying thrombocytopenia in humans and mice. Results Platelet transcriptome analyses revealed altered platelet functioning in SFTS patients and suggested an active platelet response in surviving patients but not in fatal patients. Enhanced neutrophil activation, interferon (IFN) signaling, and the virus life cycle were common platelet responses in SFTS. The increased histone methylation and impaired vesicle organization in platelets may be related to the fatal outcome, while the enhanced protein transport to membrane and RNA catabolic process may contribute to disease recovery. Moreover, SFTSV infection resulted in platelet loss via pyroptosis, apoptosis, necroptosis, and autophagy but not ferroptosis. Unlike platelets in SFTS patients, platelets in SFTSV-infected mice play a role mainly in regulating adaptive immunity, and platelet death in mice was not as severe as that in humans. Conclusions This study revealed altered platelet functioning in response to SFTSV infection and the mechanisms of thrombocytopenia in humans, which are different from those in mice infected with SFTSV. The results deepen our understanding of the pathogenesis of thrombocytopenia in SFTS and provides insights for subsequent studies on SFTS pathogenesis and the development of novel intervention strategies.
... While the hu-SGM3 model produces a higher number of megakaryocytes a precursors of blood platelets, compared with the hu-EXL, hu-NSG, and hu-NCG models, it does not appear to be suitable for the study of platelet disorders, as evidenced by their low numbers and activated profile. Since the number of megakaryocytes is higher than in the other murine models, the reduced number of human circulating platelets may be due to platelet activation and consumption [61]. However, the exact mechanism of activation remains unknown in this system. ...
Article
Full-text available
Humanized mice are an invaluable tool for investigating human diseases such as cancer, infectious diseases, and graft-versus-host disease (GvHD). However, it is crucial to understand the strengths and limitations of humanized mice and select the most appropriate model. In this study, we describe the development of the human lymphoid and myeloid lineages using a flow cytometric analysis in four humanized mouse models derived from NOD mice xenotransplanted with CD34+ fetal cord blood from a single donor. Our results showed that all murine strains sustained human immune cells within a proinflammatory environment induced by GvHD. However, the Hu-SGM3 model consistently generated higher numbers of human T cells, monocytes, dendritic cells, mast cells, and megakaryocytes, and a low number of circulating platelets showing an activated profile when compared with the other murine strains. The hu-NOG-EXL model had a similar cell development profile but a higher number of circulating platelets with an inactivated state, and the hu-NSG and hu-NCG developed low frequencies of immune cells compared with the other models. Interestingly, only the hu-SGM3 and hu-EXL models developed mast cells. In conclusion, our findings highlight the importance of selecting the appropriate humanized mouse model for specific research questions, considering the strengths and limitations of each model and the immune cell populations of interest.
... Consistent with many previous studies [8,9,21], fever, organomegaly, pancytopenia, hyperferritinemia with DIC, and hypercytokinemia are the most common clinical and laboratory findings in both sHLS/MAS and EVD [1,3]. Hypercytokinemia plays an essential role in the pathogenesis of the sHLS/MAS [22,23]. ...
Article
Background: Ebola virus disease (EVD) is one of the most severe and fatal viral hemorrhagic fevers and appears to mimic many clinical and laboratory manifestations of hemophagocytic lymphohistiocytosis syndrome (HLS), also known as macrophage activation syndrome (MAS). However, a clear association is yet to be firmly established for effective host-targeted, immunomodulatory therapeutic approaches to improve outcomes in patients with severe EVD. Methods: Twenty-four rhesus monkeys were exposed intramuscularly to the Ebola virus (EBOV) Kikwit and euthanized at prescheduled time points or when they reached the end-stage disease criteria. Three additional monkeys were mock-exposed and used as uninfected controls. Results: EBOV-exposed monkeys presented with clinicopathologic features of HLS, including fever, multi-organomegaly, pancytopenia, hemophagocytosis, hyperfibrinogenemia with disseminated intravascular coagulation, hypertriglyceridemia, hypercytokinemia, increased concentrations of soluble CD163 and CD25 in serum, and the loss of activated natural killer cells. Conclusions: Our data suggest that EVD in the rhesus macaque model mimics pathophysiologic features of HLS/MAS. Hence, regulating inflammation and immune function might provide an effective treatment for controlling acute EVD pathogenesis.