Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Adv Exp Med Biol ; 1448: 293-305, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39117823

RESUMEN

Infections caused by parasites and fungi can trigger the cytokine storm syndrome (CSS). These infections causing CSS can occur together with acquired immunodeficiencies, lymphomas, the use of immunosuppressive medications, transplant recipients, cancer, autoinflammatory, and autoimmune diseases or less frequently in healthy individuals. Histoplasma, Leishmania, Plasmodium, and Toxoplasma are the most frequent organisms associated with a CSS. It is very important to determine a previous travel history when evaluating a patient with a CSS triggered by these organisms as this may be the clue to the causal agent. Even though CSS is treated with specific therapies, an effort to find the causal organism should be carried out since the treatment of the infectious organism may stop the CSS. Diagnosing a CSS in the presence of parasitic or fungal sepsis should also lead to the study of an altered cytotoxic or hemophagocytic response in the susceptible host.


Asunto(s)
Síndrome de Liberación de Citoquinas , Humanos , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/microbiología , Micosis/microbiología , Micosis/inmunología , Animales , Enfermedades Parasitarias/inmunología , Enfermedades Parasitarias/parasitología , Enfermedades Parasitarias/complicaciones , Citocinas/metabolismo
2.
Adv Exp Med Biol ; 1448: 285-291, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39117822

RESUMEN

Zoonotic infections can result in life-threatening complications that can manifest with hemophagocytic lymphohistiocytosis (HLH)/cytokine storm syndrome (CSS). Bacteria constitute the largest group of zoonotic infection-related HLH cases. The growing list of zoonotic bacterial infections associated with HLH/CSS include Brucella spp., Rickettsia spp., Ehrlichia, Coxiella burnetii, Mycobacterium spp., and Bartonella spp. Patients most commonly present with fever, cytopenias, hepatosplenomegaly, myalgias, and less frequently with rash, jaundice, and lymphadenopathy.


Asunto(s)
Síndrome de Liberación de Citoquinas , Humanos , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/microbiología , Síndrome de Liberación de Citoquinas/etiología , Animales , Zoonosis Bacterianas/microbiología , Linfohistiocitosis Hemofagocítica/microbiología , Linfohistiocitosis Hemofagocítica/inmunología , Zoonosis/microbiología
3.
PLoS Pathog ; 18(1): e1010176, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35007290

RESUMEN

COVID-19 displays diverse disease severities and symptoms including acute systemic inflammation and hypercytokinemia, with subsequent dysregulation of immune cells. Bacterial superinfections in COVID-19 can further complicate the disease course and are associated with increased mortality. However, there is limited understanding of how SARS-CoV-2 pathogenesis and hypercytokinemia impede the innate immune function against bacterial superinfections. We assessed the influence of COVID-19 plasma hypercytokinemia on the functional responses of myeloid immune cells upon bacterial challenges from acute-phase COVID-19 patients and their corresponding recovery-phase. We show that a severe hypercytokinemia status in COVID-19 patients correlates with the development of bacterial superinfections. Neutrophils and monocytes derived from COVID-19 patients in their acute-phase showed an impaired intracellular microbicidal capacity upon bacterial challenges. The impaired microbicidal capacity was reflected by abrogated MPO and reduced NETs production in neutrophils along with reduced ROS production in both neutrophils and monocytes. Moreover, we observed a distinct pattern of cell surface receptor expression on both neutrophils and monocytes, in line with suppressed autocrine and paracrine cytokine signaling. This phenotype was characterized by a high expression of CD66b, CXCR4 and low expression of CXCR1, CXCR2 and CD15 in neutrophils and low expression of HLA-DR, CD86 and high expression of CD163 and CD11b in monocytes. Furthermore, the impaired antibacterial effector function was mediated by synergistic effect of the cytokines TNF-α, IFN-γ and IL-4. COVID-19 patients receiving dexamethasone showed a significant reduction of overall inflammatory markers in the plasma as well as exhibited an enhanced immune response towards bacterial challenge ex vivo. Finally, broad anti-inflammatory treatment was associated with a reduction in CRP, IL-6 levels as well as length of ICU stay and ventilation-days in critically ill COVID-19 patients. Our data provides insights into the transient functional dysregulation of myeloid immune cells against subsequent bacterial infections in COVID-19 patients and describe a beneficial role for the use of dexamethasone in these patients.


Asunto(s)
COVID-19/microbiología , Síndrome de Liberación de Citoquinas/complicaciones , Citocinas/metabolismo , Monocitos/virología , Neutrófilos/virología , COVID-19/virología , Síndrome de Liberación de Citoquinas/microbiología , Síndrome de Liberación de Citoquinas/virología , Humanos , Linfocitos/inmunología , Linfocitos/microbiología , Linfocitos/virología , Monocitos/inmunología , Monocitos/microbiología , Neutrófilos/inmunología , Neutrófilos/microbiología , SARS-CoV-2/patogenicidad
4.
Front Immunol ; 12: 765965, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721437

RESUMEN

Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), resulted in an unprecedented global crisis. Although primarily a respiratory illness, dysregulated immune responses may lead to multi-organ dysfunction. Prior data showed that the resident microbial communities of gastrointestinal and respiratory tracts act as modulators of local and systemic inflammatory activity (the gut-lung axis). Evolving evidence now signals an alteration in the gut microbiome, brought upon either by cytokines from the infected respiratory tract or from direct infection of the gut, or both. Dysbiosis leads to a "leaky gut". The intestinal permeability then allows access to bacterial products and toxins into the circulatory system and further exacerbates the systemic inflammatory response. In this review, we discuss the available data related to the role of the gut microbiome in the development and progression of COVID-19. We provide mechanistic insights into early data with a focus on immunological crosstalk and the microbiome's potential as a biomarker and therapeutic target.


Asunto(s)
COVID-19/microbiología , Síndrome de Liberación de Citoquinas/microbiología , Disbiosis/microbiología , Microbioma Gastrointestinal/inmunología , SARS-CoV-2/fisiología , COVID-19/inmunología , Síndrome de Liberación de Citoquinas/inmunología , Disbiosis/inmunología , Humanos , Inmunidad , Inflamación
5.
Mol Immunol ; 140: 167-174, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34717146

RESUMEN

Methicillin-resistant Staphylococcus aureus (MRSA) has become the most important pathogen of hospital-acquired (HA) or community-acquired (CA) infections. However, it is unclear of the cytokines responsible for pathological hyper-inflammation in sepsis related cytokine storm for MRSA infection. In this study, we selected typical HA-MRSA strain (YNSA163: ST239-t030-SCCmecⅢ) and two CA-MRSA isolates (YNSA7: ST59-t439-SCCmecⅣa and YNSA53: ST59-t437-SCCmecⅤb) from our previous research, infected on BALB/c mice, and analyzed the cytokine storm patterns during infection process. The animal experiments revealed the most serious lethal effect on BALB/c mice caused by YNSA7 strain infection, followed by YNSA53, and no BALB/c mice died for YNSA163 infection. Histopathological analyses revealed that lung was the most seriously damaged organs, followed by spleen and kidney, especially for CA-MRSA infection. The severe inflammatory reactions, tissue destruction, and massive exudation of inflammatory mediators and cells could be identified in CA-MRSA strains infected mice. Interleukin-6 (IL-6) and IL-10 were both highly expressed in spleen and lung of YNSA7 and YNSA53 dead cases compared with YNSA53 survived and YNSA163 cases, which demonstrated cytokine storm pattern for CA-MRSA strains infection. The results of IL-6 intervention experiment verified that the enhanced IL-6 secretion was responsible for the host lethality of YNSA7 infection. RNA-sequencing results among three MRSA isolates indicated most of the differentially expressed genes referred to cellular process, metabolism and genetic information processing of bacteria. Specifically, clpP, chp chemotaxis inhibit, fnbB, pathogencity island protein and virulence associated protein E were highly expressed in YNSA7 strain. In general, CA-MRSA strains provoked cytokine storm on BALB/c mice led to severe infection and lethality, the up-regulated of some virulence genes might play important role in pathogenesis.


Asunto(s)
Infecciones Comunitarias Adquiridas/microbiología , Síndrome de Liberación de Citoquinas/microbiología , Staphylococcus aureus Resistente a Meticilina/fisiología , Infecciones Estafilocócicas/microbiología , Animales , Infecciones Comunitarias Adquiridas/genética , Síndrome de Liberación de Citoquinas/genética , Síndrome de Liberación de Citoquinas/patología , Modelos Animales de Enfermedad , Interleucina-6/metabolismo , Estimación de Kaplan-Meier , Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Ratones Endogámicos BALB C , Especificidad de Órganos , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/patología , Transcriptoma/genética
6.
Artículo en Inglés | MEDLINE | ID: mdl-34352395

RESUMEN

p65 is one of the important subunits of the inflammation-related transcription factor NF-κB. In the present study, we cloned and identified the p65 from Megalobrama amblycephala (Mnp65) by homologous cloning and RACE technique. The full-length Mnp65 cDNA consisted of 2331 bp, and included one open reading frame encoding a 604-amino acid putative protein. The protein sequence included a DNA binding motif, a well conserved N-terminal Rel-homology domain (RHD), and a C-terminal IG-like plexins transcription (IPT). Mnp65 was closely related with the other p65 proteins of Cypriniformes and clearly distinct from that of Perciformes and Salmoniformes in terms of sequence homology. Mnp65 homodimer may interact with IκBα in the IPT domain based on the predicted 3D structure of IκBα/Mnp65 complex. Mnp65 was ubiquitously expressed in M. amblycephala tissues, and the highest levels were detected in muscle and liver. Intragastric infection with Aeromonas hydrophila caused respiratory burst and cytokine storm from 8 h to 48 h, showing significantly higher level of respiratory burst activities and significantly high cytokines levels, such as TNF-α, IL-1ß, IL-6, IL-8 etc., compared to 0 h. In addition, the bacterial challenge downregulated the IkBα, and upregulated Mnp65 and TNF-α in the liver. IkBα-Mnp65 was regulated by the negative feedback of cytokine storm, to increase IkBα and decrease Mnp65. Then cytokine storm was relieved at 96 h. Finally, severe intestinal inflammation was observed from 24 h to 48 h after infection, characterized by extensive villous necrosis, epithelial hyperplasia and lymphocyte infiltration, all of which were relieved at 96 h. Taken together, Mnp65 plays a crucial role in the physiological response of teleost fish to bacterial infection.


Asunto(s)
Aeromonas hydrophila/metabolismo , Cyprinidae/microbiología , Síndrome de Liberación de Citoquinas/inmunología , Enfermedades de los Peces/inmunología , Proteínas de Peces/metabolismo , Inflamación/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , Cyprinidae/genética , Cyprinidae/inmunología , Cyprinidae/metabolismo , Síndrome de Liberación de Citoquinas/metabolismo , Síndrome de Liberación de Citoquinas/microbiología , Síndrome de Liberación de Citoquinas/patología , Enfermedades de los Peces/metabolismo , Enfermedades de los Peces/microbiología , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Filogenia , Conformación Proteica , Estallido Respiratorio
7.
Int J Mol Sci ; 22(9)2021 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-34066560

RESUMEN

In recent decades, researchers around the world have been studying intensively how micro-organisms that are present inside living organisms could affect the main processes of life, namely health and pathological conditions of mind or body. They discovered a relationship between the whole microbial colonization and the initiation and development of different medical disorders. Besides already known probiotics, novel products such as postbiotics and paraprobiotics have been developed in recent years to create new non-viable micro-organisms or bacterial-free extracts, which can provide benefits to the host with additional bioactivity to probiotics, but without the risk of side effects. The best alternatives in the use of probiotics and postbiotics to maintain the health of the intestinal microbiota and to prevent the attachment of pathogens to children and adults are highlighted and discussed as controversies and challenges. Updated knowledge of the molecular and cellular mechanisms involved in the balance between microbiota and immune system for the introspection on the gut-lung-brain axis could reveal the latest benefits and perspectives of applied photobiomics for health. Multiple interconditioning between photobiomodulation (PBM), probiotics, and the human microbiota, their effects on the human body, and their implications for the management of viral infectious diseases is essential. Coupled complex PBM and probiotic interventions can control the microbiome, improve the activity of the immune system, and save the lives of people with immune imbalances. There is an urgent need to seek and develop innovative treatments to successfully interact with the microbiota and the human immune system in the coronavirus crisis. In the near future, photobiomics and metabolomics should be applied innovatively in the SARS-CoV-2 crisis (to study and design new therapies for COVID-19 immediately), to discover how bacteria can help us through adequate energy biostimulation to combat this pandemic, so that we can find the key to the hidden code of communication between RNA viruses, bacteria, and our body.


Asunto(s)
COVID-19/inmunología , COVID-19/microbiología , Microbioma Gastrointestinal/inmunología , Terapia por Luz de Baja Intensidad/métodos , Probióticos/uso terapéutico , SARS-CoV-2/inmunología , Encéfalo/inmunología , Encéfalo/efectos de la radiación , COVID-19/radioterapia , COVID-19/terapia , Síndrome de Liberación de Citoquinas/microbiología , Síndrome de Liberación de Citoquinas/radioterapia , Microbioma Gastrointestinal/efectos de la radiación , Humanos , Pulmón/inmunología , Pulmón/efectos de la radiación , Metabolómica , Fototerapia/métodos , SARS-CoV-2/efectos de la radiación
8.
J Neuroinflammation ; 18(1): 136, 2021 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-34130738

RESUMEN

BACKGROUND: The crucial role of type I interferon (IFN-I, IFN-α/ß) is well known to control central nervous system (CNS) neuroinflammation caused by neurotrophic flaviviruses such as Japanese encephalitis virus (JEV) and West Nile virus. However, an in-depth analysis of IFN-I signal-dependent cellular factors that govern CNS-restricted tropism in JEV infection in vivo remains to be elucidated. METHODS: Viral dissemination, tissue tropism, and cytokine production were examined in IFN-I signal-competent and -incompetent mice after JEV inoculation in tissues distal from the CNS such as the footpad. Bone marrow (BM) chimeric models were used for defining hematopoietic and tissue-resident cells in viral dissemination and tissue tropism. RESULTS: The paradoxical and interesting finding was that IFN-I signaling was essentially required for CNS neuroinflammation following JEV inoculation in distal footpad tissue. IFN-I signal-competent mice died after a prolonged neurological illness, but IFN-I signal-incompetent mice all succumbed without neurological signs. Rather, IFN-I signal-incompetent mice developed hemorrhage-like disease as evidenced by thrombocytopenia, functional injury of the liver and kidney, increased vascular leakage, and excessive cytokine production. This hemorrhage-like disease was closely associated with quick viral dissemination and impaired IFN-I innate responses before invasion of JEV into the CNS. Using bone marrow (BM) chimeric models, we found that intrinsic IFN-I signaling in tissue-resident cells in peripheral organs played a major role in inducing the hemorrhage-like disease because IFN-I signal-incompetent recipients of BM cells from IFN-I signal-competent mice showed enhanced viral dissemination, uncontrolled cytokine production, and increased vascular leakage. IFN-I signal-deficient hepatocytes and enterocytes were permissive to JEV replication with impaired induction of antiviral IFN-stimulated genes, and neuron cells derived from both IFN-I signal-competent and -incompetent mice were vulnerable to JEV replication. Finally, circulating CD11b+Ly-6C+ monocytes infiltrated into the distal tissues inoculated by JEV participated in quick viral dissemination to peripheral organs of IFN-I signal-incompetent mice at an early stage. CONCLUSION: An IFN-I signal-dependent model is proposed to demonstrate how CD11b+Ly-6C+ monocytes are involved in restricting the tissue tropism of JEV to the CNS.


Asunto(s)
Antígeno CD11b/inmunología , Virus de la Encefalitis Japonesa (Especie)/inmunología , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Monocitos/inmunología , Monocitos/microbiología , Receptor de Interferón alfa y beta , Animales , Sistema Nervioso Central/microbiología , Sistema Nervioso Central/patología , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/microbiología , Modelos Animales de Enfermedad , Virus de la Encefalitis Japonesa (Especie)/patogenicidad , Encefalitis Japonesa/inmunología , Encefalitis Japonesa/microbiología , Hemorragia/inmunología , Hemorragia/microbiología , Interacciones Huésped-Patógeno , Mediadores de Inflamación/inmunología , Tejido Linfoide/inmunología , Tejido Linfoide/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal/inmunología , Tropismo Viral
9.
Immunohorizons ; 5(6): 512-522, 2021 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-34183380

RESUMEN

Sepsis can result from a variety of pathogens, originating from a range of sources. A vast range of presenting symptoms is included in the catch-all term of "bacteremia," making diagnosis and prognosis particularly troublesome. One underexplored factor contributing to disparate outcomes is the age of the patient. Neonatal sepsis in very-low-birth-weight infants can result in vastly different immunological outcomes unique from sepsis in adults. It is also becoming increasingly clear, both from preclinical experimental models and clinical observations, that the age and history of previous microbial exposures can significantly influence the course of infection from sepsis and cytokine storms to immunopathology. In this study, we will explore key differences between neonatal and adult sepsis, experimental models used to study sepsis, and how responses to the surrounding microbial universe shape development of the immune system and impact, positively or negatively, the course of disease.


Asunto(s)
Síndrome de Liberación de Citoquinas/inmunología , Interacciones Huésped-Patógeno/inmunología , Sepsis/inmunología , Adulto , Factores de Edad , Animales , Síndrome de Liberación de Citoquinas/genética , Síndrome de Liberación de Citoquinas/microbiología , Síndrome de Liberación de Citoquinas/mortalidad , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Recién Nacido , Sepsis/genética , Sepsis/microbiología , Sepsis/mortalidad , Índice de Severidad de la Enfermedad
10.
J Cell Physiol ; 236(11): 7405-7420, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33959974

RESUMEN

Tuberculosis caused by Mycobacterium tuberculosis remains a serious global public health threat. Macrophage polarization is crucial for the innate immunity against M. tuberculosis. However, how M. tuberculosis interferes with macrophage polarization is elusive. We demonstrated here that M. tuberculosis PPE36 (Rv2108) blocked macrophage M1 polarization, preventing the cytokine storm, and alleviating inflammatory damage to mouse immune organs. PPE36 inhibited the polarization of THP-1 cell differentiation to M1 macrophages, reduced mitochondrial dehydrogenase activity, inhibited the expression of CD16, and repressed the expression of pro-inflammatory cytokines IL-6 and TNF-α, as well as chemokines CXCL9, CXCL10, CCL3, and CCL5. Intriguingly, in the mouse infection model, PPE36 significantly alleviated the inflammatory damage of immune organs caused by a cytokine storm. Furthermore, we found that PPE36 inhibited the polarization of macrophages into mature M1 macrophages by suppressing the ERK signaling. The study provided novel insights into the function and mechanism of action of M. tuberculosis effector PPE36 both at the cellular and animal level.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Síndrome de Liberación de Citoquinas/prevención & control , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Macrófagos/microbiología , Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium smegmatis/metabolismo , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/metabolismo , Síndrome de Liberación de Citoquinas/microbiología , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Interacciones Huésped-Patógeno , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Infecciones por Mycobacterium no Tuberculosas/inmunología , Infecciones por Mycobacterium no Tuberculosas/metabolismo , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/inmunología , Fenotipo , Transducción de Señal , Células THP-1
11.
J Immunother Cancer ; 9(1)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33472856

RESUMEN

BACKGROUND: Tisagenlecleucel, an anti-CD19 chimeric antigen receptor T (CAR-T) cell therapy, has demonstrated durable efficacy and a manageable safety profile in pediatric and young adult patients with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL) in the ELIANA pivotal trial and real-world experience. Experience from investigator-led studies prior to ELIANA suggests that infections and inflammatory conditions may exacerbate the severity of cytokine release syndrome (CRS) associated with CAR-T cell therapy, leading to extreme caution and strong restrictions for on-study and commercial infusion of tisagenlecleucel in patients with active infection. CRS intervention with interleukin (IL)-6 blockade and/or steroid therapy was introduced late in the course during clinical trials due to concern for potential negative effect on efficacy and persistence. However, earlier CRS intervention is now viewed more favorably. Earlier intervention and consistency in management between providers may promote broader use of tisagenlecleucel, including potential curative therapy in patients who require remission and recovery of hematopoiesis for management of severe infection. MAIN BODY: Patient 1 was diagnosed with B-ALL at 23 years old. Fourteen days before tisagenlecleucel infusion, the patient developed fever and neutropenia and was diagnosed with invasive Mucorales infection and BK virus hemorrhagic cystitis. Aggressive measures were instituted to control infection and to manage prolonged cytopenias during CAR-T cell manufacturing. Adverse events, including CRS, were manageable despite elevated inflammatory markers and active infection. The patient attained remission and recovered hematopoiesis, and infections resolved. The patient remains in remission ≥1 year postinfusion.Patient 2 was diagnosed with pre-B-ALL at preschool age. She developed severe septic shock 3 days postinitiation of lymphodepleting chemotherapy. After receiving tisagenlecleucel, she experienced CRS with cardiac dysfunction and extensive lymphadenopathy leading to renovascular compromise. The patient attained remission and was discharged in good condition to her country of origin. She remained in remission but expired on day 208 postinfusion due to cardiac arrest of unclear etiology. CONCLUSIONS: Infusion was feasible, and toxicity related to tisagenlecleucel was manageable despite active infections and concurrent inflammation, allowing attainment of remission in otherwise refractory pediatric/young adult ALL. This may lead to consideration of tisagenlecleucel as a potential curative therapy in patients with managed active infections.


Asunto(s)
Síndrome de Liberación de Citoquinas/microbiología , Inmunoterapia Adoptiva/efectos adversos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Receptores de Antígenos de Linfocitos T/administración & dosificación , Choque Séptico/microbiología , Virus BK/patogenicidad , Línea Celular Tumoral , Preescolar , Manejo de la Enfermedad , Femenino , Humanos , Bombas de Infusión , Mucormicosis/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/microbiología , Resultado del Tratamiento , Adulto Joven
12.
Cell Commun Signal ; 19(1): 7, 2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33441142

RESUMEN

The cytokine release syndrome or cytokine storm, which is the hyper-induction of inflammatory responses has a central role in the mortality rate of COVID-19 and some other viral infections. Interleukin-6 (IL-6) is a key player in the development of cytokine storms. Shedding of interleukin-6 receptor (IL-6Rα) results in the accumulation of soluble interleukin-6 receptors (sIL-6R). Only relatively few cells express membrane-bound IL-6Rα. However, sIL-6R can act on potentially all cells and organs through the ubiquitously expressed gp130, the coreceptor of IL-6Rα. Through this, so-called trans-signaling, IL-6-sIL-6R is a powerful factor in the development of cytokine storms and multiorgan involvement. Some bacteria (e.g., Serratia marcescens, Staphylococcus aureus, Pseudomonas aeruginosa, Listeria monocytogenes), commonly considered to cause co-infections during viral pneumonia, can directly induce the shedding of membrane receptors, including IL-6Rα, or enhance endogenous shedding mechanisms causing the increase of sIL-6R level. Here we hypothesise that bacteria promoting shedding and increase the sIL-6R level can be an important contributing factor for the development of cytokine storms. Therefore, inhibition of IL-6Rα shedding by drastically reducing the number of relevant bacteria may be a critical element in reducing the chance of a cytokine storm. Validation of this hypothesis can support the consideration of the prophylactic use of antibiotics more widely and at an earlier stage of infection to decrease the mortality rate of COVID-19. Video abstract.


Asunto(s)
Bacterias/enzimología , Proteínas Bacterianas/metabolismo , COVID-19/patología , Síndrome de Liberación de Citoquinas/etiología , Metaloproteasas/metabolismo , COVID-19/complicaciones , COVID-19/virología , Síndrome de Liberación de Citoquinas/microbiología , Humanos , Interleucina-6/metabolismo , Receptores de Interleucina-6/metabolismo , SARS-CoV-2/aislamiento & purificación , Transducción de Señal
13.
Toxins (Basel) ; 12(10)2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-33023185

RESUMEN

Staphylococcal superantigen toxins lead to a devastating cytokine storm resulting in shock and multi-organ failure. We have previously assessed the safety and immunogenicity of a recombinant toxic shock syndrome toxin 1 variant vaccine (rTSST-1v) in clinical trials (NCT02971670 and NCT02340338). The current study assessed neutralizing antibody titers after repeated vaccination with escalating doses of rTSST-1v. At study entry, 23 out of 34 subjects (67.6%) had neutralizing antibody titers inhibiting T cell activation as determined by 3H-thymidine incorporation at a serum dilution of ≤1:100 with similar figures for inhibition of IL-2 activation (19 of 34 subjects, 55.9%) as assessed by quantitative PCR. After the first vaccination, numbers of subjects with neutralization titers inhibiting T cell activation (61.7% ≥ 1:1000) and inhibiting IL-2 gene induction (88.2% ≥ 1:1000) increased. The immune response was augmented after the second vaccination (inhibiting T cell activation: 78.8% ≥ 1:1000; inhibiting IL-2 induction: 93.9% ≥ 1:1000) corroborated with a third immunization months later in a small subgroup of subjects. Assessment of IFNγ, TNFα and IL-6 inhibition revealed similar results, whereas neutralization titers did not change in placebo participants. Antibody titer studies show that vaccination with rTSST-1v in subjects with no/low neutralizing antibodies can rapidly induce high titer neutralizing antibodies persisting over months.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Anticuerpos Neutralizantes/sangre , Toxinas Bacterianas/administración & dosificación , Síndrome de Liberación de Citoquinas/prevención & control , Enterotoxinas/administración & dosificación , Inmunogenicidad Vacunal , Choque Séptico/prevención & control , Infecciones Estafilocócicas/prevención & control , Vacunas Estafilocócicas/administración & dosificación , Staphylococcus aureus/efectos de los fármacos , Superantígenos/administración & dosificación , Toxinas Bacterianas/genética , Toxinas Bacterianas/inmunología , Células Cultivadas , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/microbiología , Citocinas/genética , Citocinas/metabolismo , Método Doble Ciego , Enterotoxinas/genética , Enterotoxinas/inmunología , Humanos , Activación de Linfocitos/efectos de los fármacos , Estudios Prospectivos , Choque Séptico/inmunología , Choque Séptico/microbiología , Método Simple Ciego , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Vacunas Estafilocócicas/genética , Vacunas Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Staphylococcus aureus/patogenicidad , Superantígenos/genética , Superantígenos/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Tiempo , Resultado del Tratamiento , Vacunación , Vacunas Sintéticas/administración & dosificación
14.
Appl Microbiol Biotechnol ; 104(18): 7777-7785, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32780290

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a novel ß-coronavirus, is the main pathogenic agent of the rapidly spreading pneumonia called coronavirus disease 2019 (COVID-19). SARS-CoV-2 infects much more people, especially the elder population, around the world than other coronavirus, such as SARS-CoV and MERS-CoV, which is challenging current global public health system. Beyond the pathogenesis of SARS-CoV-2, microbial coinfection plays an important role in the occurrence and development of SARS-CoV-2 infection by raising the difficulties of diagnosis, treatment, prognosis of COVID-19, and even increasing the disease symptom and mortality. We summarize the coinfection of virus, bacteria and fungi with SARS-CoV-2, their effects on COVID-19, the reasons of coinfection, and the diagnosis to emphasize the importance of microbial coinfection in COVID-19. KEY POINTS: • Microbial coinfection is a nonnegligible factor in COVID-19. • Microbial coinfection exacerbates the processes of the occurrence, development and prognosis of COVID-19, and the difficulties of clinical diagnosis and treatment. • Different virus, bacteria, and fungi contributed to the coinfection with SARS-CoV-2.


Asunto(s)
Infecciones Bacterianas/epidemiología , Infecciones por Coronavirus/epidemiología , Síndrome de Liberación de Citoquinas/epidemiología , Linfopenia/epidemiología , Micosis/epidemiología , Pandemias , Neumonía Viral/epidemiología , Virosis/epidemiología , Antibacterianos/uso terapéutico , Antivirales/uso terapéutico , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/virología , Betacoronavirus/efectos de los fármacos , Betacoronavirus/inmunología , Betacoronavirus/patogenicidad , COVID-19 , Coinfección , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/microbiología , Infecciones por Coronavirus/virología , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/microbiología , Síndrome de Liberación de Citoquinas/virología , Citocinas/biosíntesis , Progresión de la Enfermedad , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Linfocitos/microbiología , Linfocitos/virología , Linfopenia/tratamiento farmacológico , Linfopenia/microbiología , Linfopenia/virología , Micosis/tratamiento farmacológico , Micosis/microbiología , Micosis/virología , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/microbiología , Neumonía Viral/virología , SARS-CoV-2 , Virosis/tratamiento farmacológico , Virosis/microbiología , Virosis/virología
15.
Virus Res ; 286: 198103, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32717345

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has evolved into a major pandemic called coronavirus disease 2019 (COVID-19) that has created unprecedented global health emergencies, and emerged as a serious threat due to its strong ability for human-to-human transmission. The reports indicate the ability of SARS-CoV-2 to affect almost any organ due to the presence of a receptor known as angiotensin converting enzyme 2 (ACE2) across the body. ACE2 receptor is majorly expressed in the brush border of gut enterocytes along with the ciliated cells and alveolar epithelial type II cells in the lungs. The amino acid transport function of ACE2 has been linked to gut microbial ecology in gastrointestinal (GI) tract, thereby suggesting that COVID-19 may, to some level, be linked to the enteric microbiota. The significant number of COVID-19 patients shows extra-pulmonary symptoms in the GI tract. Many subsequent studies revealed viral RNA of SARS-CoV-2 in fecal samples of COVID-19 patients. This presents a new challenge in the diagnosis and control of COVID-19 infection with a caution for proper sanitation and hygiene. Here, we aim to discuss the immunological co-ordination between gut and lungs that facilitates SARS-CoV-2 to infect and multiply in the inflammatory bowel disease (IBD) and non-IBD patients.


Asunto(s)
Betacoronavirus/patogenicidad , Infecciones por Coronavirus/inmunología , Síndrome de Liberación de Citoquinas/inmunología , Disbiosis/inmunología , Tracto Gastrointestinal/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Pulmón/inmunología , Neumonía Viral/inmunología , Enzima Convertidora de Angiotensina 2 , Anticuerpos Monoclonales/uso terapéutico , Antivirales/uso terapéutico , Betacoronavirus/inmunología , COVID-19 , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/microbiología , Infecciones por Coronavirus/virología , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/microbiología , Síndrome de Liberación de Citoquinas/virología , Citocinas/antagonistas & inhibidores , Citocinas/genética , Citocinas/inmunología , Disbiosis/tratamiento farmacológico , Disbiosis/microbiología , Disbiosis/virología , Microbioma Gastrointestinal/inmunología , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/virología , Expresión Génica , Interacciones Huésped-Patógeno/inmunología , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/virología , Pulmón/efectos de los fármacos , Pulmón/microbiología , Pulmón/virología , Pandemias , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/inmunología , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/microbiología , Neumonía Viral/virología , Receptores Virales/genética , Receptores Virales/inmunología , SARS-CoV-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA