Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(10)2024 May 11.
Article in English | MEDLINE | ID: mdl-38791301

ABSTRACT

Psychological stress increases risk of gastrointestinal tract diseases. However, the mechanism behind stress-induced gastrointestinal injury is not well understood. The objective of our study is to elucidate the putative mechanism of stress-induced gastrointestinal injury and develop an intervention strategy. To achieve this, we employed the restraint stress mouse model, a well-established method to study the pathophysiological changes associated with psychological stress in mice. By orally administering gut-nonabsorbable Evans blue dye and monitoring its plasma levels, we were able to track the progression of gastrointestinal injury in live mice. Additionally, flow cytometry was utilized to assess the viability, death, and inflammatory status of splenic leukocytes, providing insights into the stress-induced impact on the innate immune system associated with stress-induced gastrointestinal injury. Our findings reveal that neutrophils represent the primary innate immune leukocyte lineage responsible for stress-induced inflammation. Splenic neutrophils exhibited elevated expression levels of the pro-inflammatory cytokine IL-1, cellular reactive oxygen species, mitochondrial burden, and cell death following stress challenge compared to other innate immune cells such as macrophages, monocytes, and dendritic cells. Regulated cell death analysis indicated that NETosis is the predominant stress-induced cell death response among other analyzed regulated cell death pathways. NETosis culminates in the formation and release of neutrophil extracellular traps, which play a crucial role in modulating inflammation by binding to pathogens. Treatment with the NETosis inhibitor GSK484 rescued stress-induced neutrophil extracellular trap release and gastrointestinal injury, highlighting the involvement of neutrophil extracellular traps in stress-induced gastrointestinal inflammation. Our results suggest that neutrophil NETosis could serve as a promising drug target for managing psychological stress-induced gastrointestinal injuries.


Subject(s)
Inflammation , Neutrophils , Restraint, Physical , Stress, Psychological , Animals , Mice , Neutrophils/immunology , Neutrophils/metabolism , Stress, Psychological/complications , Stress, Psychological/immunology , Inflammation/pathology , Male , Mice, Inbred C57BL , Extracellular Traps/metabolism , Gastrointestinal Diseases/etiology , Disease Models, Animal , Reactive Oxygen Species/metabolism
2.
Tzu Chi Med J ; 36(1): 23-29, 2024.
Article in English | MEDLINE | ID: mdl-38406572

ABSTRACT

The association between stress and gastrointestinal (GI) tract diseases is well established, while the exact mechanism remains elusive. As a result, it is urgent to establish mouse models to investigate restraint stress-associated GI leakage, but current models have their limitations. A new Evans blue-fed restraint mouse model has recently been developed that allows researchers to study restraint stress-associated GI leakage in live animals. This review article will focus on this model, including its mechanisms, clinical implications, and applications for studying restraint stress-associated GI injury. Recent findings from studies using this model will also be highlighted, along with their potential for diagnosis and treatment. The article aims to discuss about current research and provide recommendations for further study, ultimately improving our understanding of the link between stress and GI injury and improving patient outcomes.

3.
Stem Cell Res Ther ; 14(1): 300, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37864264

ABSTRACT

BACKGROUND: Granulocyte colony-stimulating factor (G-CSF)-mediated mobilization of hematopoietic stem cells (HSCs) is a well-established method to prepare HSCs for transplantation nowadays. A sufficient number of HSCs is critical for successful HSC transplantation. However, approximately 2-6% of healthy stem cell donors are G-CSF-poor mobilizers for unknown reasons; thus increasing the uncertainties of HSC transplantation. The mechanism underlining G-CSF-mediated HSC mobilization remains elusive, so detailed mechanisms and an enhanced HSC mobilization strategy are urgently needed. Evidence suggests that P-selectin and P-selectin glycoprotein ligand-1 (PSGL-1) are one of the cell-cell adhesion ligand-receptor pairs for HSCs to keep contacting bone marrow (BM) stromal cells before being mobilized into circulation. This study hypothesized that blockage of PSGL-1 and P-selectin may disrupt HSC-stromal cell interaction and facilitate HSC mobilization. METHODS: The plasma levels of soluble P-selectin (sP-sel) before and after G-CSF administration in humans and male C57BL/6J mice were analyzed using enzyme-linked immunosorbent assay. Male mice with P-selectin deficiency (Selp-/-) were further employed to investigate whether P-selectin is essential for G-CSF-induced HSC mobilization and determine which cell lineage is sP-sel derived from. Finally, wild-type mice were injected with either G-CSF or recombinant sP-sel to investigate whether sP-sel alone is sufficient for inducing HSC mobilization and whether it accomplishes this by binding to HSCs and disrupting their interaction with stromal cells in the BM. RESULTS: A significant increase in plasma sP-sel levels was observed in humans and mice following G-CSF administration. Treatments of G-CSF induced a decrease in the level of HSC mobilization in Selp-/- mice compared with the wild-type (Selp+/+) controls. Additionally, the transfer of platelets derived from wild-type mice can ameliorate the defected HSC mobilization in the Selp-/- recipients. G-CSF induces the release of sP-sel from platelets, which is sufficient to mobilize BM HSCs into the circulation of mice by disrupting the PSGL-1 and P-selectin interaction between HSCs and stromal cells. These results collectively suggested that P-selectin is a critical factor for G-CSF-induced HSC mobilization. CONCLUSIONS: sP-sel was identified as a novel endogenous HSC-mobilizing agent. sP-sel injections achieved a relatively faster and more convenient regimen to mobilize HSCs in mice than G-CSF. These findings may serve as a reference for developing and optimizing human HSC mobilization in the future.


Subject(s)
Hematopoietic Stem Cell Mobilization , P-Selectin , Male , Mice , Humans , Animals , Hematopoietic Stem Cell Mobilization/methods , P-Selectin/genetics , P-Selectin/metabolism , Mice, Inbred C57BL , Hematopoietic Stem Cells/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte Colony-Stimulating Factor/metabolism , Recombinant Proteins/pharmacology
4.
Int J Mol Sci ; 24(16)2023 Aug 17.
Article in English | MEDLINE | ID: mdl-37629059

ABSTRACT

Psychological stress is widely acknowledged as a major contributor to immunosuppression, rendering individuals more susceptible to various diseases. The complex interplay between the nervous, endocrine, and immune systems underlies stress-induced immunosuppression. However, the underlying mechanisms of psychological-stress-induced immunosuppression remain unclear. In this study, we utilized a restraint stress mouse model known for its suitability in investigating physiological regulations during psychological stress. Comparing it with cold exposure, we observed markedly elevated levels of stress hormones corticosterone and cortisol in the plasma of mice subjected to restraint stress. Furthermore, restraint-stress-induced immunosuppression differed from the intravenous immunoglobulin-like immunosuppression observed in cold exposure, with restraint stress leading to increased macrophage cell death in the spleen. Suppression of pyroptosis through treatments of inflammasome inhibitors markedly ameliorated restraint-stress-induced spleen infiltration and pyroptosis cell death of macrophages in mice. These findings suggest that the macrophage pyroptosis associated with restraint stress may contribute to its immunosuppressive effects. These insights have implications for the development of treatments targeting stress-induced immunosuppression, emphasizing the need for further investigation into the underlying mechanisms.


Subject(s)
Immunosuppression Therapy , Pyroptosis , Animals , Mice , Cell Death , Macrophages , Restraint, Physical
5.
Int J Mol Sci ; 24(13)2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37446172

ABSTRACT

Metformin is one of the most commonly used drugs for type 2 diabetes mellitus. In addition to its anti-diabetic property, evidence suggests more potential applications for metformin, such as antiaging, cellular protection, and anti-inflammation. Studies have reported that metformin activates pathways with anti-inflammatory effects, enhances the integrity of gut epithelial tight junctions, and promotes a healthy gut microbiome. These actions contribute to the protective effect of metformin against gastrointestinal (GI) tract injury. However, whether metformin plays a protective role in psychological-stress-associated GI tract injury remains elusive. We aim to elucidate the potential protective effect of metformin on the GI system and develop an effective intervention strategy to counteract GI injury induced by acute psychological stress. By monitoring the levels of GI-nonabsorbable Evans blue dye in the bloodstream, we assessed the progression of GI injury in live mice. Our findings demonstrate that the administration of metformin effectively mitigated GI leakage caused by psychological stress. The GI protective effect of metformin is more potent when used on wild-type mice than on activating-transcription-factor 3 (ATF3)-deficient (ATF3-/-) mice. As such, metformin-mediated rescue was conducted in an ATF3-dependent manner. In addition, metformin-mediated protection is associated with the induction of stress-induced GI mRNA expressions of the stress-induced genes ATF3 and AMP-activated protein kinase. Furthermore, metformin treatment-mediated protection of CD326+ GI epithelial cells against stress-induced apoptotic cell death was observed in wild-type but not in ATF3-/- mice. These results suggest that metformin plays a protective role in stress-induced GI injury and that ATF3 is an essential regulator for metformin-mediated rescue of stress-induced GI tract injury.


Subject(s)
Diabetes Mellitus, Type 2 , Metformin , Mice , Animals , Activating Transcription Factor 3/genetics , Metformin/pharmacology , Epithelial Cells/metabolism , AMP-Activated Protein Kinases
6.
Int J Mol Sci ; 24(11)2023 May 25.
Article in English | MEDLINE | ID: mdl-37298220

ABSTRACT

Dengue hemorrhagic fever (DHF) is a severe form of dengue virus (DENV) infection that can lead to abnormal immune responses, endothelial vascular dysfunction, and hemorrhage pathogenesis. The virion-associated envelope protein domain III (EIII) is thought to play a role in the virulence of DENV by damaging endothelial cells. However, it is unclear whether EIII-coated nanoparticles simulating DENV virus particles could cause a more severe pathogenesis than soluble EIII alone. This study aimed to investigate whether EIII-coated silica nanoparticles (EIII-SNPs) could elicit greater cytotoxicity in endothelial cells and hemorrhage pathogenesis in mice compared to EIII or silica nanoparticles alone. The main methods included in vitro assays to assess cytotoxicity and in vivo experiments to examine hemorrhage pathogenesis in mice. EIII-SNPs induced greater endothelial cytotoxicity in vitro than EIII or silica nanoparticles alone. Two-hit combined treatment with EIII-SNPs and antiplatelet antibodies to simulate DHF hemorrhage pathogenesis during secondary DENV infections resulted in higher endothelial cytotoxicity than either treatment alone. In mouse experiments, two-hit combined treatment with EIII-SNPs and antiplatelet antibodies resulted in more severe hemorrhage pathogenesis compared to single treatments of EIII, EIII-SNPs, or antiplatelet antibodies alone. These findings suggest that EIII-coated nanoparticles are more cytotoxic than soluble EIII and could be used to develop a tentative dengue two-hit hemorrhage pathogenesis model in mice. Additionally, our results indicated that EIII-containing DENV particles could potentially exacerbate hemorrhage pathogenesis in DHF patients who have antiplatelet antibodies, highlighting the need for further research on the potential role of EIII in DHF pathogenesis.


Subject(s)
Dengue Virus , Dengue , Animals , Mice , Antibodies, Viral , Protein Domains , Endothelial Cells/metabolism , Hemorrhage/etiology
7.
Int J Mol Sci ; 23(19)2022 Oct 07.
Article in English | MEDLINE | ID: mdl-36233211

ABSTRACT

Psychological stress is associated with increased risk of gastrointestinal (GI) tract diseases. Evidence indicated that platelets facilitate GI tissue repair in intestinal anastomosis models. However, whether platelets are involved in native mechanism of the rescue of stress-induced GI injury for maintaining the GI homeostasis remains elusive. Because P-selectin-deficient (Selp-/-) mice displayed higher stress-induced GI injury compared to the wild-type (Selp+/+) mice, and P-selectin is specifically expressed in platelets, we hypothesize that P-selectin-expressing platelets play a protective role in the rescue of stress-induced GI injury. Our goal is to clarify the putative protective role of platelets in a GI system, thereby develop a feasible intervention strategy, such as platelet transfer, to overcome stress-induced GI injury. Through monitoring the plasma levels of GI-nonabsorbable Evans blue dye to reveal the progression course of GI injury in live mice, we found that intravenous treatments of purified platelets ameliorated stress-induced GI leakage. The transfer of platelets from wild-type mice was more potent than from Selp-/- mice in the rescue of stress-induced-GI leakage in the recipients. As such, platelet transfer-mediated rescue was conducted in a P-selectin dependent manner. Additionally, platelet-mediated protection is associated with corrections of stress-induced aberrant GI mRNA expressions, including tight junctions claudin 3 and occludin, as well as stress-induced genes activating transcription factor 3 and AMP-activated protein kinase, after the transfer of wild-type platelets into wild-type and Selp-/- mice. Furthermore, the stress-induced apoptosis of CD326+ GI epithelial cells was rescued by the transfer of wild type, but not P-selectin-deficient platelets. These results suggest that platelet plays a protective role for maintaining the GI homeostasis during stress in vivo, and that P-selectin is a molecular target for managing stress-induced GI tract injury.


Subject(s)
AMP-Activated Protein Kinases , Activating Transcription Factor 3 , AMP-Activated Protein Kinases/metabolism , Activating Transcription Factor 3/metabolism , Animals , Blood Platelets/metabolism , Claudin-3/metabolism , Evans Blue , Mice , Mice, Inbred C57BL , Occludin/metabolism , RNA, Messenger/metabolism
8.
Front Immunol ; 13: 806686, 2022.
Article in English | MEDLINE | ID: mdl-35444640

ABSTRACT

Nanodiamond (ND) has been developed as a carrier to conduct various in vivo diagnostic and therapeutic uses. Safety is one of the major considerations, while the hemocompatibility of ND is not clearly addressed. Here we found that, compared to the other sizes of ND with relatively inert properties, treatments of 50 nm ND induced stronger platelet aggregation, platelet pyroptosis, apoptosis and thrombocytopenia in mice. Blockage treatments of soluble P-selectin, reactive oxygen species (ROS), and Nlrp3 inflammasome inhibitors markedly suppressed such adverse effects, suggesting ND-induced platelet activation and pyroptosis involves surface P-selectin-mediated enhancement of mitochondrial superoxide levels and Nlrp3 inflammasome activation. In addition, challenges of NDs induced less platelet pyroptosis and displayed less thrombocytopenia in P-selectin (Selp-/- ), Nlrp3 (Nlrp3-/- ) and caspase-1 (Casp1-/- ) mutants, as compared to the wild type mice. Blockers of P-selectin, ROS, and Nlrp3 inflammasome pathways could be considered as antidotes for ND induced platelet activation and thrombocytopenia.


Subject(s)
Nanodiamonds , Thrombocytopenia , Animals , Apoptosis/physiology , Caspase 1/metabolism , Inflammasomes/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , P-Selectin , Platelet Aggregation , Pyroptosis , Reactive Oxygen Species/metabolism
9.
Cells ; 10(12)2021 12 14.
Article in English | MEDLINE | ID: mdl-34944038

ABSTRACT

Psychological stress increases the risk of gastrointestinal (GI) tract diseases, which involve bidirectional communication of the GI and nerves systems. Acute stress leads to GI ulcers; however, the mechanism of the native cellular protection pathway, which safeguards tissue integrality and maintains GI homeostasis, remains to be investigated. In a mouse model of this study, restraint stress induced GI leakage, abnormal tight junction protein expression, and cell death of gut epithelial cells. The expression of activating transcription factor 3 (ATF3), a stress-responsive transcription factor, is upregulated in the GI tissues of stressed animals. ATF3-deficient mice displayed an exacerbated phenotype of GI injuries. These results suggested that, in response to stress, ATF3 is part of the native cellular protective pathway in the GI system, which could be a molecular target for managing psychological stress-induced GI tract diseases.


Subject(s)
Activating Transcription Factor 3/metabolism , Gastrointestinal Diseases/etiology , Restraint, Physical , Stress, Psychological/complications , Activating Transcription Factor 3/deficiency , Animals , Caspase 3/metabolism , Duodenum/drug effects , Duodenum/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gastrointestinal Diseases/blood , Gene Expression Regulation , Mice, Inbred C57BL , Mice, Knockout , Proton Pump Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stress, Psychological/blood , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism
10.
Front Immunol ; 12: 616394, 2021.
Article in English | MEDLINE | ID: mdl-33995345

ABSTRACT

In tropical and subtropical regions, mosquito-borne dengue virus (DENV) infections can lead to severe dengue, also known as dengue hemorrhage fever, which causes bleeding, thrombocytopenia, and blood plasma leakage and increases mortality. Although DENV-induced platelet cell death was linked to disease severity, the role of responsible viral factors and the elicitation mechanism of abnormal platelet activation and cell death remain unclear. DENV and virion-surface envelope protein domain III (EIII), a cellular binding moiety of the virus particle, highly increase during the viremia stage. Our previous report suggested that exposure to such viremia EIII levels can lead to cell death of endothelial cells, neutrophils, and megakaryocytes. Here we found that both DENV and EIII could induce abnormal platelet activation and predominantly necrotic cell death pyroptosis. Blockages of EIII-induced platelet signaling using the competitive inhibitor chondroitin sulfate B or selective Nlrp3 inflammasome inhibitors OLT1177 and Z-WHED-FMK markedly ameliorated DENV- and EIII-induced thrombocytopenia, platelet activation, and cell death. These results suggest that EIII could be considered as a virulence factor of DENV, and that Nlrp3 inflammasome is a feasible target for developing therapeutic approaches against dengue-induced platelet defects.


Subject(s)
Blood Platelets/metabolism , Dengue Virus/physiology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Severe Dengue/complications , Thrombocytopenia/etiology , Thrombocytopenia/metabolism , Animals , Biomarkers , Blood Coagulation , Blood Coagulation Tests , Blood Platelets/immunology , Cell Death , Disease Models, Animal , Disease Susceptibility , Energy Metabolism , Immunophenotyping , Mice , Mice, Knockout , Mitochondria/metabolism , Platelet Activation , Protein Interaction Domains and Motifs/immunology , Severe Dengue/virology , Thrombocytopenia/blood , Thrombocytopenia/diagnosis , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism
11.
Front Immunol ; 12: 618577, 2021.
Article in English | MEDLINE | ID: mdl-33815373

ABSTRACT

Abnormal immune responses and cytokine storm are involved in the development of severe dengue, a life-threatening disease with high mortality. Dengue virus-induced neutrophil NETosis response is associated with cytokine storm; while the role of viral factors on the elicitation of excessive inflammation mains unclear. Here we found that treatments of dengue virus envelope protein domain III (EIII), cellular binding moiety of virion, is sufficient to induce neutrophil NETosis processes in vitro and in vivo. Challenges of EIII in inflammasome Nlrp3-/- and Casp1-/- mutant mice resulted in less inflammation and NETosis responses, as compared to the wild type controls. Blockages of EIII-neutrophil interaction using cell-binding competitive inhibitor or selective Nlrp3 inflammasome inhibitors OLT1177 and Z-WHED-FMK can suppress EIII-induced NETosis response. These results collectively suggest that Nlrp3 inflammsome is a molecular target for treating dengue-elicited inflammatory pathogenesis.


Subject(s)
Extracellular Traps/immunology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Interaction Domains and Motifs/immunology , Viral Envelope Proteins/immunology , Animals , Cell Line , Dengue/immunology , Dengue/metabolism , Dengue/virology , Dengue Virus/immunology , Immunophenotyping , Mice , Mice, Knockout , Mitochondria/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Recombinant Proteins , Viral Envelope Proteins/chemistry
12.
Front Immunol ; 12: 617251, 2021.
Article in English | MEDLINE | ID: mdl-33717109

ABSTRACT

Typically occurring during secondary dengue virus (DENV) infections, dengue hemorrhagic fever (DHF) causes abnormal immune responses, as well as endothelial vascular dysfunction, for which the responsible viral factor remains unclear. During peak viremia, the plasma levels of virion-associated envelope protein domain III (EIII) increases to a point at which cell death is sufficiently induced in megakaryocytes in vitro. Thus, EIII may constitute a virulence factor for endothelial damage. In this study, we examined endothelial cell death induced by treatment with DENV and EIII in vitro. Notably, pyroptosis, the major type of endothelial cell death observed, was attenuated through treatment with Nlrp3 inflammasome inhibitors. EIII injection effectively induced endothelial abnormalities, and sequential injection of EIII and DENV-NS1 autoantibodies induced further vascular damage, liver dysfunction, thrombocytopenia, and hemorrhage, which are typical manifestations in DHF. Under the same treatments, pathophysiological changes in the Nlrp3 inflammasome-deficient mice were notably reduced compared with those in the wild-type mice. These results suggest that the Nlrp3 inflammasome constitutes a potential therapeutic target for treating DENV-induced hemorrhage in DHF.


Subject(s)
Dengue Virus/physiology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Interaction Domains and Motifs/immunology , Severe Dengue/etiology , Severe Dengue/metabolism , Viral Envelope Proteins/immunology , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Apoptosis/immunology , Autoantibodies/immunology , Cytokines/metabolism , Dengue Virus/drug effects , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Nitriles/pharmacology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Recombinant Proteins/immunology , Severe Dengue/pathology
13.
Sci Rep ; 10(1): 6294, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32286343

ABSTRACT

Dengue virus (DENV) infections may cause life-threatening dengue hemorrhagic fever (DHF). Suppressed protective immunity was shown in these patients. Although several hypotheses have been formulated, the mechanism of DENV-induced immunosuppression remains unclear. Previously, we found that cross-reactive antibodies against tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor 1 (death receptor 4 [DR4]) were elicited in DHF patients, and that anti-DR4 autoantibody fractions were elicited by nonstructural protein 1 (NS1) immunizations in experimental mice. In this study, we found that anti-DR4 antibodies could suppress B lymphocyte function in vitro and in vivo. Treatment with the anti-DR4 immunoglobulin (Ig) induced caspase-dependent cell death in immortalized B lymphocyte Raji cells in vitro. Anti-DR4 Igs elicited by NS1 and DR4 immunizations markedly suppressed mouse spleen transitional T2 B (IgM+IgD+), bone marrow pre-pro-B (B220+CD43+), pre-B (B220+CD43-), and mature B cell (B220+IgD+) subsets in mice. Furthermore, functional analysis revealed that the pre-elicitation of anti-NS1 and anti-DR4 Ig titers suppressed subsequently neutralizing antibody production by immunization with DENV envelop protein. Our data suggest that the elicitation of anti-DR4 titers through DENV NS1 immunization plays a suppressive role in humoral immunity in mice.


Subject(s)
Antibodies, Viral/immunology , Immunity, Humoral , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Severe Dengue/immunology , Viral Nonstructural Proteins/immunology , Animals , Autoantibodies/blood , Cells, Cultured , Dengue Virus/immunology , Humans , Immunization , Mice , Mice, Inbred C57BL
14.
Sci Rep ; 9(1): 17497, 2019 11 25.
Article in English | MEDLINE | ID: mdl-31767905

ABSTRACT

Thrombocytopenia is usually associated with liver injury, elevated plasma aspartate aminotransferase and alanine aminotransferase levels, and high antiplatelet immunoglobulin (Ig) titers, although the mechanism behind these effects remains elusive. Deciphering the mechanism behind acute liver disease-associated thrombocytopenia may help solve difficulties in routine patient care, such as liver biopsy, antiviral therapy, and surgery. To determine whether liver damage is sufficient per se to elicit thrombocytopenia, thioacetamide (TAA)-induced hepatitis rodent models were employed. The analysis results indicated that TAA treatment transiently induced an elevation of antiplatelet antibody titer in both rats and mice. B-cell-deficient (BCD) mice, which have loss of antibody expression, exhibited markedly less thrombocytopenia and liver damage than wild-type controls. Because TAA still induces liver damage in BCD mice, this suggests that antiplatelet Ig is one of the pathogenic factors, which play exacerbating role in the acute phase of TAA-induced hepatitis. TNF-α was differentially regulated in wild-type versus BCD mice during TAA treatment, and anti-TNF treatment drastically ameliorated antiplatelet Ig induction, thrombocytopenia, and liver injury, suggesting that the TNF pathway plays a critical role in the disease progression.


Subject(s)
Antigens, Human Platelet/immunology , Autoantibodies/metabolism , Chemical and Drug Induced Liver Injury/immunology , Thioacetamide/adverse effects , Thrombocytopenia/immunology , Animals , Chemical and Drug Induced Liver Injury/complications , Disease Models, Animal , Disease Progression , Humans , Male , Mice , Rats , Thrombocytopenia/chemically induced , Tumor Necrosis Factor-alpha/metabolism
15.
Stem Cell Res Ther ; 9(1): 119, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29720275

ABSTRACT

BACKGROUND: Red blood cells are the most abundant cells in the blood that deliver oxygen to the whole body. Erythropoietin (EPO), a positive regulator of erythropoiesis, is currently the major treatment for chronic anemia. Granulocyte colony-stimulating factor (G-CSF) is a multifunctional cytokine and a well-known regulator of hematopoietic stem cell proliferation, differentiation, and mobilization. The use of EPO in combination with G-CSF has been reported to synergistically improve erythroid responses in a group of patients with myelodysplastic syndromes who did not respond to EPO treatment alone; however, the mechanism remains unclear. METHODS: C57BL/6 J mice injected with G-CSF or EPO were used to compare the erythropoiesis status and the efficiency of erythroid mobilization by flow cytometry. RESULTS: In this study, we found that G-CSF induced more orthochromatophilic erythroblast production than did EPO in the bone marrow and spleen. In addition, in contrast to EPO treatments, G-CSF treatments enhanced the efficiency of the mobilization of newly synthesized reticulocytes into peripheral blood. Our results demonstrated that the effects of G-CSF on erythropoiesis and erythrocytic mobilization were independent of EPO secretion and, in contrast to EPO, G-CSF promoted progression of erythropoiesis through transition of early stage R2 (basophilic erythroblasts) to late stage R4 (orthochromatophilic erythroblasts). CONCLUSIONS: We demonstrate for the first time that G-CSF treatments induce a faster erythropoiesis-enhancing response than that of EPO. These findings suggest an alternative approach to treating acute anemia, especially when patients are experiencing a clinical emergency in remote areas without proper blood bank supplies.


Subject(s)
Erythropoiesis/drug effects , Erythropoietin/therapeutic use , Animals , Erythropoietin/pharmacology , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL
16.
Oncotarget ; 9(16): 12781-12795, 2018 Feb 27.
Article in English | MEDLINE | ID: mdl-29560109

ABSTRACT

Mechanisms underlying cold-induced immunosuppression remain unclear. Here we found that cold exposure leads to transient receptor potential melastatin 8 (TRPM8)-dependent, renin-angiotensin-aldosterone system (RAAS)-mediated hypertension, which subsequently induces small molecule and fluid extravasation, increases plasma Ig levels, and elicits immunosuppression. An effect is similar to the clinically-used immunosuppressive treatments of intravenous immunoglobulin (IVIg) against various inflammatory diseases, such as immune thrombocytopenia (ITP). Essential roles of TRPM8 and Ig in cold-induced immunosuppression are supported by the cold-mediated amelioration of ITP and the cold-mediated suppression of bacterial clearance, which were observed in wild-type mice but not in Ig- and TRPM8-deficient mutants. Treatment with antihypertensive drugs aliskiren and losartan drastically reversed high plasma Ig levels and ameliorated cold-induced immunosuppression, indicating the involvement of the RAAS and hypertension. These results indicated that the natively increased plasma Ig level is associated with immunosuppression during periods of cold exposure, and antihypertensive drugs can be useful to manage cold-induced immunosuppression.

17.
Virulence ; 8(8): 1719-1731, 2017 11 17.
Article in English | MEDLINE | ID: mdl-28622093

ABSTRACT

Dengue virus (DENV) infection can cause severe, life-threatening events, and no specific treatments of DENV infection are currently approved. Although thrombocytopenia is frequently observed in dengue patients, its pathogenesis is still not fully understood. Previous studies have suggested that DENV-induced thrombocytopenia occurs through viral-replication-mediated megakaryopoiesis inhibition in the bone marrow; however, the exact mechanism for megakaryopoiesis suppression remains elusive. In this study, a reductionist approach was applied, in which C57B/6J mice were inoculated with recombinant DENV-envelope protein domain III (DENV-EIII) instead of the full viral particle. Our results demonstrated that DENV-EIII-suppressed megakaryopoiesis is similar to those observed with DENV infection. Furthermore, in agreement with our in vivo analyses, DENV-EIII sufficiently suppressed the megakaryopoiesis of progenitor cells from murine bone marrow and human cord blood in vitro. Additional analyses suggested that autophagy impairment and apoptosis are involved in DENV-EIII-mediated suppression of megakaryopoiesis. These data suggest that, even without viral replication, the binding of DENV-EIII to the cell surface is sufficient to suppress megakaryopoiesis.


Subject(s)
Dengue Virus/metabolism , Dengue/physiopathology , Thrombopoiesis , Viral Envelope Proteins/metabolism , Animals , Autophagy , Cell Line , Dengue/virology , Dengue Virus/chemistry , Dengue Virus/genetics , Host-Pathogen Interactions , Humans , Male , Mice , Mice, Inbred C57BL , Viral Envelope Proteins/genetics
18.
J Immunol ; 195(6): 2743-53, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26259584

ABSTRACT

The mechanisms leading to the life-threatening dengue hemorrhagic fever (DHF) remain elusive. DHF preferentially occurs during secondary dengue infections, suggesting that aberrant immune responses are involved in its development. We previously demonstrated that the autoantibodies elicited by dengue virus (DENV) nonstructural protein 1 (NS1; anti-NS1 Igs) induce plasma leakage and mortality in mice with warfarinized anticoagulant suppression. However, the involved pathogenic Ig fractions of anti-NS1 Igs remain unclear. In this study, the autoreactive Igs in patients with DHF and in NS1-immunized rabbits crossreacted with TNF-related apoptosis-inducing ligand receptor 1 (death receptor [DR]4). Challenges with the DENV in a subcytotoxic dose sensitized endothelial cells to apoptosis. Treatments with the autoantibodies induced proapoptotic activities and suppressed the surface expression of endothelial anticoagulant thrombomodulin. Combined treatments comprising the DENV and DR4 affinity-purified fractions of anti-NS1 IgGs (anti-NS1-DR4 Ig), but not preimmune control IgGs, in subcytotoxic doses led to apoptosis in endothelial cells. Treatments with the anti-NS1-DR4 Ig led to plasma leakage, coagulopathy, and morality in mice with warfarinized anticoagulant suppression. These results suggest that DR4-induced endothelial cell sensitization through NS1-elicited autoantibodies exacerbates anticoagulant suppression, vascular injury, and plasma leakage. Detecting and blocking anti-DR Igs in patients may be novel strategies for managing severe DENV infection.


Subject(s)
Autoantibodies/immunology , Dengue Virus/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Severe Dengue/pathology , Viral Nonstructural Proteins/immunology , Animals , Antibodies/immunology , Antibodies/pharmacology , Anticoagulants , Apoptosis/immunology , Blood Coagulation , Cell Line , Cell Survival , Chick Embryo , Culicidae , Endothelial Cells/immunology , Endothelial Cells/pathology , Humans , Immunoglobulin G/immunology , Mice , RNA Interference , RNA, Small Interfering , Rabbits , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Severe Dengue/immunology , Thrombomodulin/biosynthesis
19.
Thromb Haemost ; 113(5): 1060-70, 2015 May.
Article in English | MEDLINE | ID: mdl-25740324

ABSTRACT

Dengue haemorrhagic fever (DHF) typically occurs during secondary infections with dengue viruses (DENVs). Although it is generally accepted that antibody-dependent enhancement is the primary reason why patients with secondary infection are at an increased risk of developing DHF, a growing body of evidence shows that other mechanisms, such as the elicitation of antiplatelet autoantibodies by DENV nonstructural protein NS1, also play crucial roles in the pathogenesis of DHF. In this study, we developed a "two-hit" model of secondary DENV infection to examine the respective roles of DENV (first hit) and antiplatelet Igs (second hit) on the induction of haemorrhage. Mice were first exposed to DENV and then exposed to antiplatelet or anti-NS1 Igs 24 hours later. The two-hit treatment induced substantial haemorrhage, coagulopathy, and cytokine surge, and additional treatment with antagonists of TNF-α, IL-1, caspase-1, and FcγRIII ameliorated such effects. In addition, knockout mice lacking the Fcγ receptor III, Toll-like receptor 3, and inflammasome components Nlrp3 and caspase-1 exhibited considerably fewer pathological alterations than did wild type controls. These findings may provide new perspectives for developing feasible approaches to treat patients with DHF.


Subject(s)
Antibodies, Viral/immunology , Carrier Proteins/immunology , Dengue Virus/pathogenicity , Hemorrhage/immunology , Inflammasomes/immunology , Receptors, IgG/immunology , Animals , Antibodies, Viral/blood , Autoantibodies/immunology , Female , Hemorrhage/virology , Interleukin-1/metabolism , Leukocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Recombinant Proteins/immunology , Severe Dengue/immunology , Severe Dengue/virology , Thromboplastin/metabolism , Toll-Like Receptor 3/metabolism
20.
J Biomed Sci ; 19: 98, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23170801

ABSTRACT

BACKGROUND: Lethal toxin (LT) is a major virulence factor of Bacillus anthracis. Sprague Dawley rats manifest pronounced lung edema and shock after LT treatments, resulting in high mortality. The heart failure that is induced by LT has been suggested to be a principal mechanism of lung edema and mortality in rodents. Since LT-induced death occurs more rapidly in rats than in mice, suggesting that other mechanisms in addition to the heart dysfunction may be contributed to the fast progression of LT-induced pathogenesis in rats. Coagulopathy may contribute to circulatory failure and lung injury. However, the effect of LT on coagulation-induced lung dysfunction is unclear. METHODS: To investigate the involvement of coagulopathy in LT-mediated pathogenesis, the mortality, lung histology and coagulant levels of LT-treated rats were examined. The effects of activated protein C (aPC) on LT-mediated pathogenesis were also evaluated. RESULTS: Fibrin depositions were detected in the lungs of LT-treated rats, indicating that coagulation was activated. Increased levels of plasma D-dimer and thrombomodulin, and the ameliorative effect of aPC further suggested that the activation of coagulation-fibrinolysis pathways plays a role in LT-mediated pathogenesis in rats. Reduced mortality was associated with decreased plasma levels of D-dimer and thrombomodulin following aPC treatments in rats with LT-mediated pathogenesis. CONCLUSIONS: These findings suggest that the activation of coagulation in lung tissue contributes to mortality in LT-mediated pathogenesis in rats. In addition, anticoagulant aPC may help to develop a feasible therapeutic strategy.


Subject(s)
Anthrax , Bacillus anthracis , Heart Failure/chemically induced , Protein C , Animals , Anthrax/drug therapy , Anthrax/microbiology , Anthrax/physiopathology , Antigens, Bacterial/drug effects , Antigens, Bacterial/toxicity , Bacillus anthracis/drug effects , Bacillus anthracis/pathogenicity , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/toxicity , Blood Coagulation/drug effects , Heart Failure/drug therapy , Heart Failure/metabolism , Heart Failure/physiopathology , Humans , Injections, Intraventricular , Lung/blood supply , Lung/drug effects , Lung/metabolism , Lung/physiopathology , Protein C/administration & dosage , Protein C/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...