Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Int J Mol Sci ; 23(6)2022 Mar 21.
Article in English | MEDLINE | ID: covidwho-1765735

ABSTRACT

B-cell lymphoma and lymphoproliferative diseases represent a heterogeneous and complex group of neoplasms that are accompanied by a broad range of immune regulatory disorder phenotypes. Clinical features of autoimmunity, hyperinflammation, immunodeficiency and infection can variously dominate, depending on the immune pathway most involved. Immunological imbalance can play a role in lymphomagenesis, also supporting the progression of the disease, while on the other hand, lymphoma acts on the immune system to weaken immunosurveillance and facilitate immunoevasion. Therefore, the modulation of immunity can have a profound effect on disease progression or resolution, which makes the immune system a critical target for new therapies. In the current therapeutic scenario enriched by chemo-free regimens, it is important to establish the effect of various drugs on the disease, as well as on the restoration of immune functions. In fact, treatment of B-cell lymphoma with passive immunotherapy that targets tumor cells or targets the tumor microenvironment, together with adoptive immunotherapy, is becoming more frequent. The aim of this review is to report relevant data on the evolution of the immune system during and after treatment with targeted therapy of B-cell lymphomas.


Subject(s)
Lymphoma, B-Cell , Lymphoma , Lymphoproliferative Disorders , Humans , Immune System/pathology , Immunotherapy, Adoptive , Lymphoma/drug therapy , Lymphoma, B-Cell/therapy , Lymphoproliferative Disorders/pathology , Tumor Microenvironment
2.
Cells ; 11(4)2022 02 10.
Article in English | MEDLINE | ID: covidwho-1690345

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is an extremely contagious disease whereby the virus damages the host's respiratory tract via entering through the ACE2 receptor. Cardiovascular disorder is being recognized in the majority of COVID-19 patients; yet, the relationship between SARS-CoV-2 and heart failure has not been established. In the present study, SARS-CoV-2 infection was induced in the monkey model. Thereafter, heart tissue samples were collected, and pathological changes were analyzed in the left ventricular tissue by hematoxylin and eosin, trichrome, and immunohistochemical staining specific to T lymphocytes and macrophages. The findings revealed that SARS-CoV-2 infection induces several pathological changes in the heart, which cause cardiomyocyte disarray, mononuclear infiltrates of inflammatory cells, and hypertrophy. Furthermore, collagen-specific staining showed the development of cardiac fibrosis in the interstitial and perivascular regions in the hearts of infected primates. Moreover, the myocardial tissue samples displayed multiple foci of inflammatory cells positive for T lymphocytes and macrophages within the myocardium. These findings suggest the progression of the disease, which can lead to the development of severe complications, including heart failure. Additionally, SARS-CoV-2 antigen staining detected the presence of virus particles in the myocardium. Thus, we found that SARS-CoV-2 infection is characterized by an exaggerated inflammatory immune response in the heart, which possibly contributes to myocardial remodeling and subsequent fibrosis.


Subject(s)
COVID-19/immunology , Heart Failure/physiopathology , Heart/physiopathology , Animals , Chlorocebus aethiops , Heart/virology , Heart Failure/virology , Heart Ventricles/physiopathology , Heart Ventricles/virology , Immune System/pathology , Macaca mulatta , Myocarditis/virology , Myocardium/metabolism , SARS-CoV-2/pathogenicity
3.
Autoimmunity ; 54(4): 213-224, 2021 06.
Article in English | MEDLINE | ID: covidwho-1201340

ABSTRACT

Currently, the novel coronavirus pneumonia has been widespread globally, and there is no specific medicine. In response to the emergency, we employed bioinformatics methods to investigate the virus's pathogenic mechanism, finding possible control methods. We speculated in previous studies that E protein was associated with viral infectivity. The present study adopted the domain search techniques to analyse the E protein. According to the results, the E protein could bind iron or haem. The iron and haem bound by the E protein came from the attacked haemoglobin and phagocytes. When E protein was attached to haem, it synthesised oxygen and water into superoxide anions, hydrogen peroxide and hydroxyl radicals. When the iron-bound E protein and the haem-bound E protein worked together, they converted superoxide anions and hydrogen peroxide into oxygen and water. These were the "ROS attack" and "ROS escape" of the virus. "ROS attack" damaged the tissues or cells exposed on the surface of the virus, and "ROS escape" decomposed the superoxide anion and hydrogen peroxide that attacked the virus. When NK cells were exposed to infected cells, viruses that had not shed from the infected cells' surface damaged them through "ROS attack". In addition, lymphocytes such as T cells and B cells, which could be close to the antigen of the virus surface, were also easily damaged or killed by the "ROS attack", generating a decrease in lymphocytes. When memory B cells were exposed to the virus's surface antigen, they were also damaged by "ROS attack", resulting in the patient's re-infection. The virus applied the "ROS escape" to decompose hydrogen peroxide released by phagocytes into oxygen and water. The surrounding cells were replenished with oxygen, and the patient was in a "happy hypoxia" state. When the phagocytes swallowed the virus, the E protein converted superoxide anions into oxygen and water. In this way, the virus parasitized in the vesicles of the phagocyte. While virus was in the lysosome, the E protein generated ROS to damage nearby hydrolases. In this way, the virus parasitized the lysosome. Excessive hydroxyl free radicals destroyed the membrane structure of the lysosome, causing the hydrolase release from lysosome, autophagy of phagocytic cells and subsequent cell death. As a result, the colonizing phagocytes of the virus was associated with asymptomatic infection or retest-positive. Briefly, the virus inhibited the immune system through "ROS escape", and damaged the immune system by "ROS attack". The destruction instigated a strong cytokine storm, leading to organ failure and complications.


Subject(s)
COVID-19/etiology , COVID-19/metabolism , Disease Susceptibility , Host-Pathogen Interactions , Immune System/immunology , Immune System/metabolism , Iron/metabolism , Reactive Oxygen Species/metabolism , SARS-CoV-2/physiology , Amino Acid Sequence , Catalysis , Computational Biology/methods , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immune System/pathology , Models, Molecular , Protein Conformation , Structure-Activity Relationship , Superoxide Dismutase/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism
4.
Front Endocrinol (Lausanne) ; 12: 596518, 2021.
Article in English | MEDLINE | ID: covidwho-1156116

ABSTRACT

Clinical Trial Registration: www.ClinicalTrials.gov, identifier: NCT04365634. Context: Diabetes mellitus was associated with increased severity and mortality of disease in COVID-19 pneumonia. So far the effect of type 2 diabetes (T2DM) or hyperglycemia on the immune system among COVID-19 disease has remained unclear. Objective: We aim to explore the clinical and immunological features of type 2 diabetes mellitus (T2DM) among COVID-19 patients. Design and Methods: In this retrospective study, the clinical and immunological characteristics of 306 hospitalized confirmed COVID-19 patients (including 129 diabetic and 177 non-diabetic patients) were analyzed. The serum concentrations of laboratory parameters including cytokines and numbers of immune cells were measured and compared between diabetic and non-diabetic groups. Results: Compared with non-diabetic group, diabetic cases more frequently had lymphopenia and hyperglycemia, with higher levels of urea nitrogen, myoglobin, D-dimer and ferritin. Diabetic cases indicated the obviously elevated mortality and the higher levels of cytokines IL-2R, IL-6, IL-8, IL-10, and TNF-α, as well as the distinctly reduced Th1/Th2 cytokines ratios compared with non-diabetic cases. The longitudinal assays showed that compared to that at week 1, the levels of IL-6 and IL-8 were significantly elevated at week 2 after admission in non-survivors of diabetic cases, whereas there were greatly reductions from week 1 to week 2 in survivors of diabetic cases. Compared with survival diabetic patients, non-survival diabetic cases displayed distinct higher serum concentrations of IL-2R, IL-6, IL-8, IL-10, TNF-α, and lower Th1/Th2 cytokines ratios at week 2. Samples from a subset of participants were evaluated by flow cytometry for the immune cells. The counts of peripheral total T lymphocytes, CD4+ T cells, CD8+ T cells and NK cells were markedly lower in diabetic cases than in non-diabetic cases. The non-survivors showed the markedly declined counts of CD8+ T cells and NK cells than survivors. Conclusion: The elevated cytokines, imbalance of Th1/Th2 cytokines ratios and reduced of peripheral numbers of CD8+ T cells and NK cells might contribute to the pathogenic mechanisms of high mortality of COVID-19 patients with T2DM.


Subject(s)
COVID-19/immunology , Diabetes Mellitus, Type 2/immunology , Adult , Aged , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , COVID-19/blood , COVID-19/complications , COVID-19/mortality , China/epidemiology , Cytokines/analysis , Cytokines/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/mortality , Female , Humans , Hyperglycemia/blood , Hyperglycemia/complications , Hyperglycemia/immunology , Hyperglycemia/mortality , Immune System/metabolism , Immune System/pathology , Killer Cells, Natural/pathology , Lymphocyte Count , Lymphopenia/blood , Lymphopenia/complications , Lymphopenia/immunology , Lymphopenia/mortality , Male , Middle Aged , Retrospective Studies , SARS-CoV-2/immunology , SARS-CoV-2/physiology , Th1 Cells/pathology , Th2 Cells/pathology
5.
Nat Biotechnol ; 38(8): 970-979, 2020 08.
Article in English | MEDLINE | ID: covidwho-1023942

ABSTRACT

To investigate the immune response and mechanisms associated with severe coronavirus disease 2019 (COVID-19), we performed single-cell RNA sequencing on nasopharyngeal and bronchial samples from 19 clinically well-characterized patients with moderate or critical disease and from five healthy controls. We identified airway epithelial cell types and states vulnerable to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In patients with COVID-19, epithelial cells showed an average three-fold increase in expression of the SARS-CoV-2 entry receptor ACE2, which correlated with interferon signals by immune cells. Compared to moderate cases, critical cases exhibited stronger interactions between epithelial and immune cells, as indicated by ligand-receptor expression profiles, and activated immune cells, including inflammatory macrophages expressing CCL2, CCL3, CCL20, CXCL1, CXCL3, CXCL10, IL8, IL1B and TNF. The transcriptional differences in critical cases compared to moderate cases likely contribute to clinical observations of heightened inflammatory tissue damage, lung injury and respiratory failure. Our data suggest that pharmacologic inhibition of the CCR1 and/or CCR5 pathways might suppress immune hyperactivation in critical COVID-19.


Subject(s)
Coronavirus Infections/pathology , Coronavirus Infections/physiopathology , Pneumonia, Viral/pathology , Pneumonia, Viral/physiopathology , Respiratory System/pathology , Single-Cell Analysis , Transcriptome , Adult , Aged , Angiotensin-Converting Enzyme 2 , Bronchoalveolar Lavage Fluid/virology , COVID-19 , Cell Communication , Cell Differentiation , Coronavirus Infections/virology , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Humans , Immune System/pathology , Inflammation/immunology , Inflammation/pathology , Longitudinal Studies , Male , Middle Aged , Nasopharynx/virology , Pandemics , Peptidyl-Dipeptidase A/genetics , Pneumonia, Viral/virology , Respiratory System/immunology , Respiratory System/virology , Severity of Illness Index
6.
J Transl Med ; 18(1): 415, 2020 11 07.
Article in English | MEDLINE | ID: covidwho-916349

ABSTRACT

On December 12, 2019 a new coronavirus (SARS-CoV-2) emerged in Wuhan, China, triggering a pandemic of severe acute respiratory syndrome in humans (COVID-19). Today, the scientific community is investing all the resources available to find any therapy and prevention strategies to defeat COVID-19. In this context, immunonutrition can play a pivotal role in improving immune responses against viral infections. Immunonutrition has been based on the concept that malnutrition impairs immune function. Therefore, immunonutrition involves feeding enriched with various pharmaconutrients (Omega 3 Fatty Acids, Vitamin C, Arginine, Glutamine, Selenium, Zinc, Vitamin, E and Vitamin D) to modulate inflammatory responses, acquired immune response and to improve patient outcomes. In literature, significant evidences indicate that obesity, a malnutrition state, negatively impacts on immune system functionality and on host defense, impairing protection from infections. Immunonutrients can promote patient recovery by inhibiting inflammatory responses and regulating immune function. Immune system dysfunction is considered to increase the risk of viral infections, such as SARS-CoV-2, and was observed in different pathological situations. Obese patients develop severe COVID-19 sequelae, due to the high concentrations of TNF-α, MCP-1 and IL-6 produced in the meantime by visceral and subcutaneous adipose tissue and by innate immunity. Moreover, leptin, released by adipose tissue, helps to increase inflammatory milieu with a dysregulation of the immune response. Additionally, gut microbiota plays a crucial role in the maturation, development and functions of both innate and adaptive immune system, as well as contributing to develop obese phenotype. The gut microbiota has been shown to affect lung health through a vital crosstalk between gut microbiota and lungs, called the "gut-lung axis". This axis communicates through a bi-directional pathway in which endotoxins, or microbial metabolites, may affect the lung through the blood and when inflammation occurs in the lung, this in turn can affect the gut microbiota. Therefore, the modulation of gut microbiota in obese COVID-19 patients can play a key role in immunonutrition therapeutic strategy. This umbrella review seeks to answer the question of whether a nutritional approach can be used to enhance the immune system's response to obesity in obese patients affected by COVID-19.


Subject(s)
Betacoronavirus/physiology , Coronavirus Infections/complications , Coronavirus Infections/immunology , Immune System/pathology , Immune System/virology , Nutritional Physiological Phenomena , Obesity/complications , Obesity/virology , Pneumonia, Viral/complications , Pneumonia, Viral/immunology , COVID-19 , Coronavirus Infections/microbiology , Humans , Microbiota , Obesity/microbiology , Pandemics , Pneumonia, Viral/microbiology , SARS-CoV-2
7.
Int J Immunopathol Pharmacol ; 34: 2058738420966497, 2020.
Article in English | MEDLINE | ID: covidwho-881050

ABSTRACT

Coronavirus disease 2019 (COVID-19), an acute respiratory infection, is largely associated with dysregulation and impairment of the immune system. This study investigated how the immune system changes were related to disease severity in COVID-19 patients. The frequencies of different immune cells and levels of pro- and anti-inflammatory cytokines in whole blood of participants were determined by flow cytometry and enzyme-linked immunosorbent assay, respectively. The values of other inflammatory agents were also studied. In the late recovery stage, unlike CD56high CD16+/- NK cells and monocytes, CD56low CD16+ NK cell numbers were increased (P < 0.0001-0.05). Th1, Th2, and Th17 cell percentages were significantly lower in patients than healthy control (P < 0.0001-0.05), while their frequencies were increased following disease recovery (P < 0.0001-0.05). The numbers of Tregs, activated CD4+ T cells, and exhausted CD8+ T cells were significantly decreased during a recovery (P < 0.0001-0.05). No significant change was observed in exhausted CD4+ T cell number during a recovery (P > 0.05). B cell showed an increased percentage in patients compared to healthy subjects (P < 0.0001-0.05), whereas its number was reduced following recovery (P < 0.0001-0.05). IL-1α, IL-1ß, IL-6, TNF-α, and IL-10 levels were significantly decreased in the late recovery stage (P < 0.0001-0.05). However, TGF-ß1 level was not significantly changed during the recovery (P > 0.05). Lymphocyte numbers in patients were significantly decreased (P < 0.001), unlike ESR value (P < 0.001). Lymphocyte number was negatively correlated to ESR value and Th2 number (P < 0.05), while its association with monocyte was significantly positive at the first day of recovery (P < 0.05). The immune system changes during the disease recovery to improve and regulate immune responses and thereby may associate with the reduction in disease severity.


Subject(s)
Coronavirus Infections/immunology , Coronavirus Infections/pathology , Immune System/immunology , Immune System/pathology , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Aged , Aged, 80 and over , Blood Sedimentation , COVID-19 , Case-Control Studies , Cytokines/metabolism , Female , Flow Cytometry , Humans , Leukocyte Count , Lymphocyte Count , Male , Middle Aged , Pandemics , Recovery of Function , T-Lymphocytes/immunology , Treatment Outcome
8.
Neuroscientist ; 27(4): 331-339, 2021 08.
Article in English | MEDLINE | ID: covidwho-797803

ABSTRACT

The coronavirus disease 2019 (COVID-19) is a pandemic disease, originated in Wuhan City, China. It is caused by severe acute respiratory syndrome corona virus 2 (SARS-CoV-2) and its biology is still poorly understood. Currently, there are no vaccines and drugs/or agents that can reduce severity of this new disease. Recent data suggest that patients with age-related comorbidities, including cardiovascular disease, diabetes, obesity, hypertension, chronic kidney disease, and dementia are highly susceptible to severe respiratory illness due to coronavirus infection. Recent research also revealed that aged individuals with elevated baseline inflammation cause defects in T and B cells, leading to decreased body's immune response to viral infection. In the current article, we discuss the effects of SARS-CoV-2 on age-related chronic diseases, such as diabetes, obesity, and Alzheimer's disease. Our article also highlights the interaction between coronavirus and immune cells, and how COVID-19 alters mitochondrial activities in host cells. Based on new and compelling evidence, we propose that mitochondrial fission is inhibited while fusion is promoted, causing mitochondrial elongation and providing a receptive intracellular environment for viral replication in infected cells. Further research is still needed to understand the cross talk between viral replication in mitochondria and disease progression in patients with COVID-19.


Subject(s)
COVID-19/immunology , COVID-19/pathology , Dementia/immunology , Dementia/pathology , Diabetes Complications/immunology , Diabetes Complications/pathology , Diabetes Mellitus/immunology , Diabetes Mellitus/pathology , Immune System/pathology , Mitochondria/pathology , Mitochondrial Dynamics , Obesity/immunology , Obesity/pathology , Humans , Immunity, Cellular
9.
Immunol Cell Biol ; 99(2): 192-205, 2021 02.
Article in English | MEDLINE | ID: covidwho-735921

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic has affected the daily lives of millions of people worldwide and had caused significant mortality; hence, the assessment of therapeutic options is of great interest. The leading cause of death among COVID-19 patients is acute respiratory distress syndrome caused by hyperinflammation secondary to cytokine release syndrome (CRS). Cytokines, such as tumor necrosis factor-α, interleukin-6, interferon-γ and interleukin-10, are the main mediators of CRS. Based on recent evidence, the angiotensin-converting enzyme (ACE) II is known to be the target of the COVID-19 spike protein, which enables the virus to penetrate human cells. ACE II also possesses an anti-inflammatory role in many pathologies such as cardiovascular disease, hypertension, diabetes mellitus and other conditions, which are the main risk factors of poor prognosis in COVID-19 infection. Changes in tissue ACE II levels are associated with many diseases and hyperinflammatory states, and it is assumed that elevated levels of ACE II could aggravate the course of COVID-19 infection. Therefore, the use of renin-angiotensin-aldosterone system inhibitors (RASis) in COVID-19 patients could be hypothetically considered, though sufficient evidence is not presented by the scientific community. In this work, based on the most recent pieces of evidence, the roles of RAS and RASi in immunologic interactions are addressed. Furthermore, the molecular and immunologic aspects of RASi and their potential significance in COVID-19 are discussed.


Subject(s)
Angiotensin-Converting Enzyme 2/physiology , COVID-19 , SARS-CoV-2/physiology , Virus Internalization , Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme 2/antagonists & inhibitors , Angiotensin-Converting Enzyme 2/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , COVID-19/immunology , COVID-19/pathology , COVID-19/virology , Humans , Immune System/metabolism , Immune System/pathology , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , SARS-CoV-2/drug effects , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Virus Internalization/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL