Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 744
Filter
1.
FASEB J ; 38(7): e23597, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38581235

ABSTRACT

Sepsis is a life-threatening condition that occurs when the body responds to an infection but subsequently triggers widespread inflammation and impaired blood flow. These pathologic responses can rapidly cause multiple organ dysfunction or failure either one by one or simultaneously. The fundamental common mechanisms involved in sepsis-induced multiple organ dysfunction remain unclear. Here, employing quantitative global and phosphoproteomics, we examine the liver's temporal proteome and phosphoproteome changes after moderate sepsis induced by cecum ligation and puncture. In total, 4593 global proteins and 1186 phosphoproteins according to 3275 phosphosites were identified. To characterize the liver-kidney comorbidity after sepsis, we developed a mathematical model and performed cross-analyses of liver and kidney proteome data obtained from the same set of mice. Beyond immune response, we showed the commonly disturbed pathways and key regulators of the liver-kidney comorbidity are linked to energy metabolism and consumption. Our data provide open resources to understand the communication between the liver and kidney as they work to fight infection and maintain homeostasis.


Subject(s)
Proteome , Sepsis , Mice , Animals , Multiple Organ Failure/complications , Multiple Organ Failure/pathology , Liver/metabolism , Kidney/metabolism , Sepsis/metabolism , Disease Models, Animal
2.
Sci Total Environ ; 894: 164842, 2023 Oct 10.
Article in English | MEDLINE | ID: mdl-37336398

ABSTRACT

N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine quinone (6-PPDQ) is the ozonation product of tire antioxidant 6-PPD. 6-PPDQ can be detected in different environments, such as roadway runoff and dust. Although 6-PPDQ toxicity has been frequently assessed in aquatic organisms, the possible toxic effects of 6-PPDQ on mammals remain largely unclear. We here aimed to perform systematic assessment to evaluate 6-PPDQ toxicity on multiple organs in mice. Male BALB/c mice were intraperitoneally injected with 6-PPDQ for two exposure modes, single intraperitoneal injection and repeated intraperitoneal injection every four days for 28 days. Serum, liver, kidney, lung, spleen, testis, brain, and heart were collected for injury evaluation by organ index, histopathology analysis and biochemical parameters. In 0.4 and 4 mg/kg 6-PPDQ single injected mice, no significant changes in organ indexes and biochemical parameters were detected, and only moderate pathological changes were observed in organs of liver, kidney, lung, and brain. Very different from this, in 0.4 and 4 mg/kg 6-PPDQ repeated injected mice, we observed the obvious increase in organ indexes of liver, kidney, lung, testis, and brain, and the decrease in spleen index. Meanwhile, the significant pathological changes were formed in liver, kidney, lung, spleen, testis, and brain in 0.4 and 4 mg/kg 6-PPDQ repeated injected mice. Biochemical parameters of liver (alanine aminotransferase (ALT), aspartate aminotransferase (AST), and alkaline phosphatase (ALP)) and kidney (urea and creatinine) were all significantly upregulated by repeated injection with 0.4 and 4 mg/kg 6-PPDQ. After repeated exposure, most of 6-PPDQ was accumulated in liver and lung of mice. Therefore, our results suggested the risk of repeated exposure to 6-PPDQ in inducing toxicity on multiple organs in mice.


Subject(s)
Antioxidants , Benzoquinones , Multiple Organ Failure , Phenylenediamines , Animals , Male , Mice , Antioxidants/toxicity , Kidney/drug effects , Kidney/pathology , Liver/drug effects , Liver/pathology , Mice, Inbred BALB C , Phenylenediamines/toxicity , Benzoquinones/toxicity , Multiple Organ Failure/chemically induced , Multiple Organ Failure/pathology
3.
Acta Cardiol ; 78(8): 863-877, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37318070

ABSTRACT

The signalling mechanisms involving the kidney and heart are a niche of networks causing pathological conditions inducing inflammation, reactive oxidative species, cell apoptosis, and organ dysfunction during the onset of clinical complications. The clinical manifestation of the kidney and heart depends on various biochemical processes that influence organ dysfunction coexistence through circulatory networks, which hold utmost importance. The cells of both organs also influence remote communication, and evidence states that it may be explicitly by circulatory small noncoding RNAs, i.e. microRNAs (miRNAs). Recent developments target miRNAs as marker panels for disease diagnosis and prognosis. Circulatory miRNAs expressed in renal and cardiac disease can reveal relevant information about the niche of networks and gene transcription and regulated networks. In this review, we discuss the pertinent roles of identified circulatory miRNAs regulating signal transduction pathways critical in the onset of renal and cardiac disease, which can hold promising future targets for clinical diagnostic and prognostic purposes.


Subject(s)
Heart Diseases , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Multiple Organ Failure/pathology , Kidney , Heart Diseases/diagnosis , Heart Diseases/genetics , Heart Diseases/metabolism
4.
Cell ; 186(9): 1824-1845, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37116469

ABSTRACT

Cachexia, a systemic wasting condition, is considered a late consequence of diseases, including cancer, organ failure, or infections, and contributes to significant morbidity and mortality. The induction process and mechanistic progression of cachexia are incompletely understood. Refocusing academic efforts away from advanced cachexia to the etiology of cachexia may enable discoveries of new therapeutic approaches. Here, we review drivers, mechanisms, organismal predispositions, evidence for multi-organ interaction, model systems, clinical research, trials, and care provision from early onset to late cachexia. Evidence is emerging that distinct inflammatory, metabolic, and neuro-modulatory drivers can initiate processes that ultimately converge on advanced cachexia.


Subject(s)
Cachexia , Humans , Cachexia/drug therapy , Cachexia/etiology , Cachexia/metabolism , Cachexia/pathology , Muscle, Skeletal/metabolism , Neoplasms/complications , Neoplasms/metabolism , Neoplasms/pathology , Infections/complications , Infections/pathology , Multiple Organ Failure/complications , Multiple Organ Failure/pathology
5.
Radiat Res ; 199(4): 319-335, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36857032

ABSTRACT

The objective of the current study was to establish a mouse model of acute radiation syndrome (ARS) after total-body irradiation with 2.5% bone marrow sparing (TBI/BM2.5) that progressed to the delayed effects of acute radiation exposure, specifically pneumonitis and/or pulmonary fibrosis (DEARE-lung), in animals surviving longer than 60 days. Two hundred age and sex matched C57L/J mice were assigned to one of six arms to receive a dose of 9.5 to 13.25 Gy of 320 kV X-ray TBI/BM2.5. A sham-irradiated cohort was included as an age- and sex-matched control. Blood was sampled from the facial vein prior to irradiation and on days 5, 10, 15, 20, 25, and 30 postirradiation for hematology. Respiratory function was monitored at regular intervals throughout the in-life phase. Animals with respiratory dysfunction were administered a single 12-day tapered regimen of dexamethasone, allometrically scaled from a similar regimen in the non-human primate. All animals were monitored daily for up to 224 days postirradiation for signs of organ dysfunction and morbidity/mortality. At euthanasia due to criteria or at the study endpoint, wet lung weights were recorded, and blood sampled for hematology and serum chemistry. The left lung, heart, spleen, small and large intestine, and kidneys were processed for histopathology. A dose-response curve with the estimated lethal dose for 10-99% of animals with 95% confidence intervals was established. The median survival time was significantly prolonged in males as compared to females across the 10.25 to 12.5 Gy dose range. Animal sex played a significant role in overall survival, with males 50% less likely to expire prior to the study endpoint compared to females. All animals developed pancytopenia within the first one- to two-weeks after TBI/BM2.5 followed by a progressive recovery through day 30. Fourteen percent of animals expired during the first 30-days postirradiation due to ARS (e.g., myelosuppression, gastrointestinal tissue abnormalities), with most deaths occurring prior to day 15. Microscopic findings show the presence of radiation pneumonitis as early as day 57. At time points later than day 70, pneumonitis was consistently present in the lungs of mice and the severity was comparable across radiation dose arms. Pulmonary fibrosis was first noted at day 64 but was not consistently present and stable in severity until after day 70. Fibrosis was comparable across radiation dose arms. In conclusion, this study established a multiple organ injury mouse model that progresses through the ARS phase to DEARE-lung, characterized by respiratory dysfunction, and microscopic abnormalities consistent with radiation pneumonitis/fibrosis. The model provides a platform for future development of medical countermeasures for approval and licensure by the U.S. Food and Drug Administration under the animal rule regulatory pathway.


Subject(s)
Pneumonia , Pulmonary Fibrosis , Radiation Pneumonitis , United States , Male , Animals , Female , Mice , Bone Marrow/radiation effects , Radiation Pneumonitis/pathology , Multiple Organ Failure/pathology , Disease Models, Animal , Mice, Inbred Strains , Fibrosis
6.
Pol Merkur Lekarski ; 51(6): 608-612, 2023.
Article in English | MEDLINE | ID: mdl-38207061

ABSTRACT

OBJECTIVE: Aim: Using quantitative morphological methods to study the peculiarities of the structural reconstruction of the venous bad of the prostate at the conditions of post-resection portal hypertension. PATIENTS AND METHODS: Materials and Methods: Morphologically, the venous bed of the prostate of 15 intact white rats, 30 animals with post-resection portal hypertension, 17 rats with a combination of post-resection portal hypertension with hepatargia, enteral, cardiac, and renal insufficiency was studied. Rats were slaughtered one month after the start of the experiment by bloodletting under general thiopental anesthesia. Morphometry of the venous blood vessels of the prostate gland was performed on histological specimens, during which the diameter of the postcapillary venules, the diameter of the venules, the external diameter of the venous vessels, the internal diameter of the venous vessels, the thickness of the wall of the venous vessels was determined. Also it was studied the height of endothelial cells, diameter of their nuclei, nuclear-cytoplasmic ratio in these cells, relative volume of damaged endothelial cells, density of vessels of microcirculatory bed per 1 mm² of prostate tissue. RESULTS: Results: It was found that resection of the left and right lobes of the liver leads to the development of postresection portal hypertension and pronounced remodeling of the vessels of the venous bed of the prostate gland, which was characterized by the expansion of the capillary venules of the prostate by 36.5%, with the occurrence of multiple organ failure by 38.5% (р<0.001 ), an increase in the lumen of veins, thinning of their wall, atrophy, dystrophy, and necrobiosis of endothelial cells, disruption of structural cellular homeostasis, endothelial dysfunction, hypoxia, dystrophic-necrotic changes in cells, stromal structures, infiltration, and sclerosis. Morphological changes in the structures of the prostate dominated when post-resection portal hypertension was combined with multiple organ failure. CONCLUSION: Conclusions: Post-resection portal hypertension in laboratory sexually mature white male rats leads to pronounced remodeling of the venous bed of the prostate gland, which is characterized by the expansion of the lumen of the vessels, thinning of their walls, atrophic, dystrophic and necrobiotic changes in endothelial cells, a violation of structural cellular homeostasis in them, endothelial dysfunction, hypoxia, dystrophic-necrotic changes in cells, stromal structures, infiltration and sclerosis. Morphological changes in the structures of the prostate dominated when post-resection portal hypertension was combined with multiple organ failure.


Subject(s)
Hypertension, Portal , Prostate , Male , Rats , Animals , Prostate/surgery , Prostate/pathology , Microcirculation , Endothelial Cells/pathology , Sclerosis/pathology , Multiple Organ Failure/pathology , Hypertension, Portal/etiology , Hypertension, Portal/pathology , Hypoxia/pathology
7.
Biomolecules ; 12(12)2022 12 07.
Article in English | MEDLINE | ID: mdl-36551258

ABSTRACT

Leptospirosis is an important zoonotic disease, causing about 60,000 deaths annually. In this review, we have described in detail the immunopathogenesis of leptospirosis, the influence of cytokines, genetic susceptibility on the course of the disease, and the evasion of the immune response. These data are combined with information about immunological and pathomorphological changes in the kidneys, liver, and lungs, which are most affected by Weil's disease. The review also suggests a possible role of the gut microbiota in the clinical course of leptospirosis, the main mechanisms of the influence of gut dysbiosis on damage in the liver, kidneys, and lungs through several axes, i.e., gut-liver, gut-kidney, and gut-lungs. Modulation of gut microbiota by probiotics and/or fecal microbiota transplantation in leptospirosis may become an important area of scientific research.


Subject(s)
Gastrointestinal Microbiome , Leptospirosis , Weil Disease , Humans , Weil Disease/pathology , Multiple Organ Failure/pathology , Liver/pathology
8.
Elife ; 112022 09 30.
Article in English | MEDLINE | ID: mdl-36178806

ABSTRACT

Sepsis is a life-threatening condition characterized by uncontrolled systemic inflammation and coagulation, leading to multiorgan failure. Therapeutic options to prevent sepsis-associated immunopathology remain scarce. Here, we established a mouse model of long-lasting disease tolerance during severe sepsis, manifested by diminished immunothrombosis and organ damage in spite of a high pathogen burden. We found that both neutrophils and B cells emerged as key regulators of tissue integrity. Enduring changes in the transcriptional profile of neutrophils include upregulated Cxcr4 expression in protected, tolerant hosts. Neutrophil Cxcr4 upregulation required the presence of B cells, suggesting that B cells promoted disease tolerance by improving tissue damage control via the suppression of neutrophils' tissue-damaging properties. Finally, therapeutic administration of a Cxcr4 agonist successfully promoted tissue damage control and prevented liver damage during sepsis. Our findings highlight the importance of a critical B-cell/neutrophil interaction during sepsis and establish neutrophil Cxcr4 activation as a potential means to promote disease tolerance during sepsis.


Subject(s)
Bacterial Infections , Sepsis , Animals , Bacterial Infections/metabolism , Disease Models, Animal , Mice , Multiple Organ Failure/metabolism , Multiple Organ Failure/pathology , Neutrophils/metabolism , Sepsis/metabolism
9.
Biomed Pharmacother ; 148: 112768, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35247717

ABSTRACT

Pulmonary fibrosis induced by silica particles is defined as silicosis, which is an incurable disease. The pathogenesis of silicosis is not completely clear, but it's certain that immune system dysfunction is closely related to it. Immune checkpoint inhibitors (ICIs) are emerging immunotherapeutic agents that mainly target adaptive immune cells, and there is abundant evidence that ICIs are of great value in cancer treatment. However, whether these attractive agents can be implemented in silicosis treatment is unclear. In this study, we explored the efficacy of small molecule inhibitors targeted PD-1/PD-L1 and CTLA-4 on silica-induced pulmonary fibrosis in mice. ICIs were injected intraperitoneally into mice that received silica instillation twice a week. The mice were sacrificed 7 and 28 days after the injection. The lungs, spleen, hilar lymph nodes, thymus, and peripheral blood of mice were collected and subjected to histological examination, flow cytometry analysis, and mRNA and protein quantification. Our results demonstrated that silica exposure caused damage to multiple immune organs in mice, leading to an imbalance in systemic immune homeostasis. Specifically, proportions and subtypes of T and B cells were significantly altered, and the expressions of PD-1, PD-L1 and CTLA-4 were abnormal on these cells. Both PD-1/PD-L1 and CTLA-4 inhibitor administration modulated silica-induced immune system disruption, however, only PD-1/PD-L1 signaling inhibition showed significant amelioration of silicosis. Our findings confirmed for the first time the potential value of ICIs for the treatment of silica-induced pulmonary fibrosis, and this may provide new ideas for the treatment of other fibrosis-related diseases.


Subject(s)
Immune Checkpoint Inhibitors/pharmacology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Silicon Dioxide/adverse effects , Animals , B-Lymphocyte Subsets/drug effects , B7-H1 Antigen/drug effects , CTLA-4 Antigen/drug effects , Homeostasis/drug effects , Male , Mice , Mice, Inbred C57BL , Multiple Organ Failure/chemically induced , Multiple Organ Failure/pathology , Programmed Cell Death 1 Receptor/drug effects , RNA, Messenger , T-Lymphocyte Subsets/drug effects
10.
Acta Pharmacol Sin ; 43(3): 520-528, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34040166

ABSTRACT

High mobility group box 1 (HMGB1) is a ubiquitous nuclear protein that is present in almost all cells and regulates the activity of innate immune responses in both intracellular and extracellular settings. Current evidence suggests that HMGB1 plays a pivotal role in human pathological and pathophysiological processes such as the inflammatory response, immune reactions, cell migration, aging, and cell death. Sepsis is a systemic inflammatory response syndrome (SIRS) that occurs in hosts in response to microbial infections with a proven or suspected infectious etiology and is the leading cause of death in intensive care units worldwide, particularly in the aging population. Dysregulated systemic inflammation is a classic characteristic of sepsis, and suppression of HMGB1 may ameliorate inflammation and improve patient outcomes. Here, we focus on the latest breakthroughs regarding the roles of HMGB1 in sepsis and sepsis-related organ injury, the ways by which HMGB1 are released, and the signaling pathways and therapeutics associated with HMGB1. This review highlights recent advances related to HMGB1: the regulation of HMBG1 might be helpful for both basic research and drug development for the treatment of sepsis and sepsis-related organ injury.


Subject(s)
HMGB1 Protein/metabolism , Multiple Organ Failure/pathology , Sepsis/pathology , Autophagy/physiology , Blood Coagulation Disorders/pathology , Cytokine Release Syndrome/pathology , Endoplasmic Reticulum Stress/physiology , Humans , Inflammation/pathology , Inflammation Mediators/metabolism , Mitochondria/pathology , Multiple Organ Failure/drug therapy , Receptor for Advanced Glycation End Products/metabolism , Sepsis/drug therapy , Signal Transduction/physiology , Toll-Like Receptors/metabolism
11.
J Clin Pathol ; 75(6): 383-389, 2022 Jun.
Article in English | MEDLINE | ID: mdl-33722841

ABSTRACT

BACKGROUND: Secondary haemophagocytic lymphohistiocytosis (sHLH) is characterised by a hyper activation of immune system that leads to multiorgan failure. It is suggested that excessive immune response in patients with COVID-19 could mimic this syndrome. Some COVID-19 autopsy studies have revealed the presence of haemophagocytosis images in bone marrow, raising the possibility, along with HScore parameters, of sHLH. AIM: Our objective is to ascertain the existence of sHLH in some patients with severe COVID-19. METHODS: We report the autopsy histological findings of 16 patients with COVID-19, focusing on the presence of haemophagocytosis in bone marrow, obtained from rib squeeze and integrating these findings with HScore parameters. CD68 immunohistochemical stains were used to highlight histiocytes and haemophagocytic cells. Clinical evolution and laboratory parameters of patients were collected from electronic clinical records. RESULTS: Eleven patients (68.7%) displayed moderate histiocytic hyperplasia with haemophagocytosis (HHH) in bone marrow, three patients (18.7%) displayed severe HHH and the remainder were mild. All HScore parameters were collected in 10 patients (62.5%). Among the patients in which all parameters were evaluable, eight patients (80%) had an HScore >169. sHLH was not clinically suspected in any case. CONCLUSIONS: Our results support the recommendation of some authors to use the HScore in patients with severe COVID-19 in order to identify those who could benefit from immunosuppressive therapies. The presence of haemophagocytosis in bone marrow tissue, despite not being a specific finding, has proved to be a very useful tool in our study to identify these patients.


Subject(s)
COVID-19 , Lymphohistiocytosis, Hemophagocytic , Autopsy , Bone Marrow/pathology , COVID-19/complications , Humans , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/pathology , Multiple Organ Failure/pathology
12.
Respiration ; 101(2): 155-165, 2022.
Article in English | MEDLINE | ID: mdl-34525475

ABSTRACT

BACKGROUND: Findings from autopsies have provided evidence on systemic microvascular damage as one of the underlying mechanisms of Coronavirus disease 2019 (CO-VID-19). The aim of this study was to correlate autopsy-based cause of death in SARS-CoV-2, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) positive patients with chest imaging and severity grade of pulmonary and systemic morphological vascular pathology. METHODS: Fifteen SARS-CoV-2 positive autopsies with clinically distinct presentations (age 22-89 years) were retrospectively analyzed with focus on vascular, thromboembolic, and ischemic changes in pulmonary and in extrapulmonary sites. Eight patients died due to COVID-19 associated respiratory failure with diffuse alveolar damage in various stages and/or multi-organ failure, whereas other reasons such as cardiac decompensation, complication of malignant tumors, or septic shock were the cause of death in 7 further patients. The severity of gross and histopathological changes was semi-quantitatively scored as 0 (absent), 1 (mild), and 3 (severe). Severity scores between the 2 groups were correlated with selected clinical parameters, initial chest imaging, autopsy-based cause of death, and compared using Pearson χ2 and Mann-Whitney U tests. RESULTS: Severe pulmonary endotheliitis (p = 0.031, p = 0.029) and multi-organ involvement (p = 0.026, p = 0.006) correlated significantly with COVID-19 associated death. Pulmonary microthrombi showed limited statistical correlation, while tissue necrosis, gross pulmonary embolism, and bacterial superinfection did not differentiate the 2 study groups. Chest imaging at hospital admission did not differ either. CONCLUSIONS: Extensive pulmonary endotheliitis and multi-organ involvement are characteristic autopsy features in fatal CO-VID-19 associated deaths. Thromboembolic and ischemic events and bacterial superinfections occur frequently in SARS-CoV-2 infection independently of outcome.


Subject(s)
COVID-19/mortality , COVID-19/pathology , Endothelium, Vascular/pathology , Multiple Organ Failure/virology , Respiratory Distress Syndrome/virology , Vasculitis/virology , Adult , Aged , Aged, 80 and over , Autopsy , COVID-19/complications , Cause of Death , Cohort Studies , Female , Humans , Male , Middle Aged , Multiple Organ Failure/mortality , Multiple Organ Failure/pathology , Pulmonary Alveoli/pathology , Respiratory Distress Syndrome/mortality , Respiratory Distress Syndrome/pathology , Vasculitis/mortality , Vasculitis/pathology , Young Adult
13.
Biomed Pharmacother ; 146: 112503, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34922113

ABSTRACT

Cancer is the second most common cause of death in the United States and is a challenging disease to treat. The treatment options for various cancers include but are not limited to surgery, radiation, and chemotherapy. The mechanism behind chemotherapy is intended to promote cellular damage to cells that are proliferating uncontrollably. Unfortunately for the recipients, most chemotherapeutic agents cannot differentiate between malignant cells and healthy cells and tissues. Thus, chemotherapy-induced toxicities are often observed in once-healthy organs. These effects can be acute and self-limiting or chronic, appearing long after chemotherapy is completed. Cancer survivors can then present for non-cancer related surgeries later in life, due to this toxicity. Furthermore, the administration of chemotherapeutic agents can profoundly impact the anesthetic management of patients who are undergoing surgery. This review discusses how chemotherapy-induced organ toxicity can occur in multiple organ systems and what drugs should be avoided if prior toxicity exists in these organ systems.


Subject(s)
Antineoplastic Agents/adverse effects , Multiple Organ Failure/chemically induced , Multiple Organ Failure/pathology , Neoplasms/drug therapy , Perioperative Care/methods , Anesthesia/methods , Anesthetics/therapeutic use , Antineoplastic Agents/therapeutic use , Clinical Protocols , Humans , Multiple Organ Failure/prevention & control , Pain, Postoperative/drug therapy , Surgical Procedures, Operative/methods
14.
Viral Immunol ; 34(10): 679-688, 2021 12.
Article in English | MEDLINE | ID: mdl-34882013

ABSTRACT

The newfound coronavirus disease 2019 (COVID-19), initiated by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is an international public health concern, threatening the lives of millions of people worldwide. The virus seems to have a propensity to infect older males, especially those with underlying diseases. The cytokine storm following hyperactivated immune responses due to SARS-CoV-2 infection is probably the crucial source of severe pneumonia that leads to acute lung injury, systemic inflammatory response syndrome, or acute respiratory distress syndrome, and finally multiple organ dysfunction syndromes, as well as death in many cases. Several studies revealed that interleukin (IL)-1ß levels were elevated during COVID-19 infection. In addition, the IL-1 cytokine family has a pivotal role in the induction of cytokine storm due to uncontrolled immune responses in COVID-19 infection. This article reviews the role of IL-1 in inflammation and utilization of IL-1 inhibitor agents in controlling the inflammatory outcomes initiated by SARS-CoV-2 infection.


Subject(s)
COVID-19 Drug Treatment , COVID-19/immunology , Cytokine Release Syndrome/drug therapy , Interleukin-1/immunology , Acute Lung Injury/drug therapy , Acute Lung Injury/immunology , Acute Lung Injury/pathology , COVID-19/mortality , COVID-19/pathology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/pathology , Humans , Interleukin-1/antagonists & inhibitors , Multiple Organ Failure/drug therapy , Multiple Organ Failure/immunology , Multiple Organ Failure/pathology , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/pathology , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity
15.
Clin Appl Thromb Hemost ; 27: 10760296211051764, 2021.
Article in English | MEDLINE | ID: mdl-34755565

ABSTRACT

The precise mechanisms of pathology in severe COVID-19 remains elusive. Current evidence suggests that inflammatory mediators are responsible for the manifestation of clinical symptoms that precedes a fatal response to infection. This review examines the nature of platelet activating factor and emphasizes the similarities between the physiological effects of platelet activating factor and the clinical complications of severe COVID-19.


Subject(s)
COVID-19/metabolism , Platelet Activating Factor/metabolism , Animals , COVID-19/complications , COVID-19/mortality , COVID-19/pathology , Humans , Inflammation/complications , Inflammation/metabolism , Inflammation/mortality , Inflammation/pathology , Multiple Organ Failure/complications , Multiple Organ Failure/metabolism , Multiple Organ Failure/mortality , Multiple Organ Failure/pathology , Respiratory Distress Syndrome/complications , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/mortality , Respiratory Distress Syndrome/pathology , SARS-CoV-2/physiology , Severity of Illness Index , Thrombosis/complications , Thrombosis/metabolism , Thrombosis/mortality , Thrombosis/pathology
16.
Cell Death Dis ; 12(11): 1041, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34725342

ABSTRACT

The regulation of intracellular calcium (Ca2+) homeostasis is fundamental to maintain normal functions in many cell types. The ryanodine receptor (RyR), the largest intracellular calcium release channel located on the sarco/endoplasmic reticulum (SR/ER), plays a key role in the intracellular Ca2+ handling. Abnormal type 2 ryanodine receptor (RyR2) function, associated to mutations (ryanopathies) or pathological remodeling, has been reported, not only in cardiac diseases, but also in neuronal and pancreatic disorders. While animal models and in vitro studies provided valuable contributions to our knowledge on RyR2 dysfunctions, the human cell models derived from patients' cells offer new hope for improving our understanding of human clinical diseases and enrich the development of great medical advances. We here discuss the current knowledge on RyR2 dysfunctions associated with mutations and post-translational remodeling. We then reviewed the novel human cellular technologies allowing the correlation of patient's genome with their cellular environment and providing approaches for personalized RyR-targeted therapeutics.


Subject(s)
Disease , Models, Biological , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Humans , Induced Pluripotent Stem Cells/metabolism , Multiple Organ Failure/pathology , Mutation/genetics , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/genetics
17.
Medicine (Baltimore) ; 100(46): e27891, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34797336

ABSTRACT

RATIONALE: Studies have previously reported misidentifying Caltha palustris (C. palustris) as Ligularia fischeri and its subsequent ingestion leading to abdominal pain and gastrointestinal symptoms, which are alleviated immediately. Bradycardia and hypotension may persist for several days, and an infusion of dopamine can restore a healthy state without complications. We report a case of C. palustris poisoning with protein-losing enteropathy that has not been reported previously. The patient died of multiple organ failure, and exhibited more severe clinical deterioration than previous cases due to prolonged shock. PATIENT CONCERNS: A 70-year-old woman was admitted to the emergency department (ED) with complaints of epigastric pain, vomiting, and diarrhea after ingestion of a poisonous plant presumed to be C. palustris. The patient presented with bradycardia and hypotension after ED admission, and vasopressor infusion improved bradycardia but not hypotension, while the patient complained of severe epigastric pain. DIAGNOSES: Abdominal computed tomography showed luminal distention and edematous thickening of the entire stomach lining, as well as small and large intestinal wall edema, indicating severe gastritis and enterocolitis. The laboratory test results suggested severe hypoalbuminemia, while the arterial blood gas analyses showed a continuous increase in metabolic acidosis. INTERVENTIONS: As plant poisoning was suspected, activated charcoal was administered to the patient, followed by administration of vasopressors and other conservative therapies. Continuous renal replacement therapy (CRRT) was used for metabolic acidosis of increasing severity. OUTCOMES: Despite the administration of vasopressors and other conservative therapies, the state of shock persisted, and metabolic acidosis did not improve even after CRRT. Ultimately, the patient died of multiple organ failure. LESSONS: For many poisonous wild plants, the precise profile of toxic compounds and mechanisms of action remain to be identified; when there is insufficient literature reporting on suspected plant poisoning, the medical personnel providing the treatment should consider the various side effects that differ from the reported ones and the possibility of more severe clinical progress and poor prognosis.


Subject(s)
Acidosis , Multiple Organ Failure/chemically induced , Nausea/chemically induced , Plant Poisoning/diagnosis , Vomiting/chemically induced , Abdominal Pain/etiology , Aged , Bradycardia , Eating , Fatal Outcome , Female , Humans , Hypotension/chemically induced , Multiple Organ Failure/pathology , Plant Poisoning/complications , Vasoconstrictor Agents , Vomiting/diagnosis
18.
Sci Rep ; 11(1): 22772, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34815465

ABSTRACT

We hypothesized that the composition of sepsis-inducing bacterial flora influences the course of fecal peritonitis in rodents. Saline or fecal suspensions with a standardized dose range of bacterial colony-forming units (CFUs) were injected intraperitoneally into Sprague-Dawley rats. The qualitative composition of the initial inoculum and the ascites was analyzed separately by MALDI-TOF mass spectrometry. Invasive monitoring was conducted in separate anesthetized groups (n = 12-13/group) after 12, 24, 48 and 72 h to determine rat-specific organ failure assessment (ROFA) scores. Death and ROFA scores peaked at 24 h. At this time, 20% mortality occurred in animals receiving a monomicrobial E. coli suspension, and ROFA scores were significantly higher in the monomicrobial subgroup than in the polymicrobial one (median 6.5; 5.0-7.0 and 5.0; 4.75-5.0, respectively). ROFA scores dropped after 48 h, accompanied by a steady decrease in ascites CFUs and a shift towards intra-abdominal monomicrobial E. coli cultures. Furthermore, we found a relationship between ascites CFUs and the evolving change in ROFA scores throughout the study. Hence, quantitatively identical bacterial loads with mono- or polymicrobial dominance lead to a different degree of sepsis severity and divergent outcomes. Initial and intraperitoneal microbiological testing should be used to improve translational research success.


Subject(s)
Bacteria/classification , Bacteria/pathogenicity , Bacterial Infections/complications , Disease Models, Animal , Multiple Organ Failure/pathology , Sepsis/pathology , Animals , Bacteria/isolation & purification , Bacterial Infections/microbiology , Male , Multiple Organ Failure/etiology , Prognosis , Rats , Rats, Sprague-Dawley , Sepsis/etiology , Sepsis/microbiology
19.
Front Immunol ; 12: 742941, 2021.
Article in English | MEDLINE | ID: mdl-34659238

ABSTRACT

The coronavirus disease-19 (COVID-19) elicited by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused devastating health, economic and social impact worldwide. Its clinical spectrum ranges from asymptomatic to respiratory failure and multi-organ failure or death. The pathogenesis of SARS-CoV-2 infection is attributed to a complex interplay between virus and host immune response. It involves activation of multiple inflammatory pathways leading to hyperinflammation and cytokine storm, resulting in tissue damage, acute respiratory distress syndrome (ARDS) and multi-organ failure. Accumulating evidence has raised concern over the long-term health effects of COVID-19. Importantly, the neuroinvasive potential of SARS-CoV-2 may have devastating consequences in the brain. This review provides a conceptual framework on how the virus tricks the host immune system to induce infection and cause severe disease. We also explore the key differences between mild and severe COVID-19 and its short- and long-term effects, particularly on the human brain.


Subject(s)
Adaptive Immunity/immunology , COVID-19/pathology , Cytokine Release Syndrome/pathology , Immunity, Innate/immunology , SARS-CoV-2/immunology , COVID-19/complications , COVID-19/immunology , Cytokine Release Syndrome/immunology , Cytokines/blood , Humans , Multiple Organ Failure/pathology , Respiratory Distress Syndrome/pathology , Sex Factors , Post-Acute COVID-19 Syndrome
20.
Sci Rep ; 11(1): 17772, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34493741

ABSTRACT

We investigated the impact of aerobic exercise (AE) on multiple organ dysfunction syndrome (MODS), aortic injury, pathoglycemia, and death during sepsis. ICR mice were randomized into four groups: Control (Con), Lipopolysaccharide (LPS), Exercise (Ex), and Exercise + LPS (Ex + LPS) groups. Mice were trained with low-intensity for 4 weeks. LPS and Ex + LPS mice received 5 mg/kg LPS intraperitoneally for induction of sepsis. Histopathological micrographs showed the organ morphology and damage. This study examined the effects of AE on LPS-induced changes in systemic inflammation, pulmonary inflammation, lung permeability, and bronchoalveolar lavage fluid (BALF) cell count, oxidative stress-related indicators in the lung, blood glucose levels, plasma lactate levels, serum insulin levels, plasma high-mobility group box 1 (HMGB1) levels, glucose transporter 1 (Glut1) and HMGB1, silent information regulator 1 (Sirt-1), and nuclear factor erythroid 2-related factor 2 (Nrf-2) mRNA expression levels in lung tissue. AE improved sepsis-associated multiple organ dysfunction syndrome (MODS), aortic injury, hypoglycemia, and death. AE prominently decreased pulmonary inflammation, pulmonary edema, and modulated redox balance during sepsis. AE prominently decreased neutrophil content in organ. AE prominently downregulated CXCL-1, CXCL-8, IL-6, TNF-α, Glu1, and HMGB1 mRNA expression but activated IL-1RN, IL-10, Sirt-1, and Nrf-2 mRNA expression in the lung during sepsis. AE decreased the serum levels of lactate and HMGB1 but increased blood glucose levels and serum insulin levels during sepsis. A 4-week AE improves sepsis-associated MODS, aortic injury, pathoglycemia, and death. AE impairs LPS-induced lactate and HMGB1 release partly because AE increases serum insulin levels and decreases the levels of Glut1. AE is a novel therapeutic strategy for sepsis targeting aerobic glycolysis.


Subject(s)
Endotoxemia/therapy , Exercise , Glycolysis/physiology , Multiple Organ Failure/prevention & control , Physical Conditioning, Animal/physiology , Animals , Blood Glucose/analysis , Bronchoalveolar Lavage Fluid/cytology , Cytokines/analysis , Endotoxemia/chemically induced , Endotoxemia/complications , Glucose Transporter Type 1/blood , HMGB1 Protein/blood , Humans , Immunity, Innate , Insulin/blood , Lactates/blood , Lipopolysaccharides/toxicity , Lung/pathology , Male , Mice , Mice, Inbred ICR , Multiple Organ Failure/etiology , Multiple Organ Failure/immunology , Multiple Organ Failure/pathology , Neutrophils/pathology , Oxidative Stress , Random Allocation , Viscera/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...