Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Front Immunol ; 14: 1162739, 2023.
Article in English | MEDLINE | ID: covidwho-2314172

ABSTRACT

Cytokines are secretion proteins that mediate and regulate immunity and inflammation. They are crucial in the progress of acute inflammatory diseases and autoimmunity. In fact, the inhibition of proinflammatory cytokines has been widely tested in the treatment of rheumatoid arthritis (RA). Some of these inhibitors have been used in the treatment of COVID-19 patients to improve survival rates. However, controlling the extent of inflammation with cytokine inhibitors is still a challenge because these molecules are redundant and pleiotropic. Here we review a novel therapeutic approach based on the use of the HSP60-derived Altered Peptide Ligand (APL) designed for RA and repositioned for the treatment of COVID-19 patients with hyperinflammation. HSP60 is a molecular chaperone found in all cells. It is involved in a wide diversity of cellular events including protein folding and trafficking. HSP60 concentration increases during cellular stress, for example inflammation. This protein has a dual role in immunity. Some HSP60-derived soluble epitopes induce inflammation, while others are immunoregulatory. Our HSP60-derived APL decreases the concentration of cytokines and induces the increase of FOXP3+ regulatory T cells (Treg) in various experimental systems. Furthermore, it decreases several cytokines and soluble mediators that are raised in RA, as well as decreases the excessive inflammatory response induced by SARS-CoV-2. This approach can be extended to other inflammatory diseases.


Subject(s)
Arthritis, Rheumatoid , Chaperonin 60 , Humans , COVID-19 , Cytokines/metabolism , Inflammation/drug therapy , Peptides/pharmacology , Peptides/therapeutic use , SARS-CoV-2/metabolism , Chaperonin 60/pharmacology , Chaperonin 60/therapeutic use
2.
Int J Mol Sci ; 23(17)2022 Sep 04.
Article in English | MEDLINE | ID: covidwho-2010115

ABSTRACT

Background: Hyperinflammation is frequently associated with the chronic pain of autoimmune disease and the acute death of coronavirus disease (COVID-19) via a severe cytokine cascade. CIGB-258 (Jusvinza®), an altered peptide ligand with 3 kDa from heat shock protein 60 (HSP60), inhibits the systemic inflammation and cytokine storm, but the precise mechanism is still unknown. Objective: The protective effect of CIGB-258 against inflammatory stress of N-ε-carboxymethyllysine (CML) was tested to provide mechanistic insight. Methods: CIGB-258 was treated to high-density lipoproteins (HDL) and injected into zebrafish and its embryo to test a putative anti-inflammatory activity under presence of CML. Results: Treatment of CML (final 200 µM) caused remarkable glycation of HDL with severe aggregation of HDL particles to produce dysfunctional HDL, which is associated with a decrease in apolipoprotein A-I stability and lowered paraoxonase activity. Degradation of HDL3 by ferrous ions was attenuated by a co-treatment with CIGB-258 with a red-shift of the Trp fluorescence in HDL. A microinjection of CML (500 ng) into zebrafish embryos resulted in the highest embryo death rate, only 18% of survivability with developmental defects. However, co-injection of CIGB-258 (final 1 ng) caused the remarkable elevation of survivability around 58%, as well as normal developmental speed. An intraperitoneal injection of CML (final 250 µg) into adult zebrafish resulted acute paralysis, sudden death, and laying down on the bottom of the cage with no swimming ability via neurotoxicity and inflammation. However, a co-injection of CIGB-258 (1 µg) resulted in faster recovery of the swimming ability and higher survivability than CML alone injection. The CML alone group showed 49% survivability, while the CIGB-258 group showed 97% survivability (p < 0.001) with a remarkable decrease in hepatic inflammation up to 50%. A comparison of efficacy with CIGB-258, Infliximab (Remsima®), and Tocilizumab (Actemra®) showed that the CIGB-258 group exhibited faster recovery and swimming ability with higher survivability than those of the Infliximab group. The CIGB-258 group and Tocilizumab group showed the highest survivability, the lowest plasma total cholesterol and triglyceride level, and the infiltration of inflammatory cells, such as neutrophils in hepatic tissue. Conclusion: CIGB-258 ameliorated the acute neurotoxicity, paralysis, hyperinflammation, and death induced by CML, resulting in higher survivability in zebrafish and its embryos by enhancing the HDL structure and functionality.


Subject(s)
COVID-19 , Lipoproteins, HDL , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Inflammation/drug therapy , Infliximab , Lysine/analogs & derivatives , Paralysis , Zebrafish/metabolism
3.
Revista Cubana de Medicina Militar ; 50(4), 2021.
Article in Spanish | Scopus | ID: covidwho-1801644

ABSTRACT

Introduction: Infection with SARS-CoV-2 induces a prothrombotic state in patients, by the combination of hyperinflammatory response and hypoxia. In Cuba, the drug called Jusvinza, based on an immunomodulatory peptide, is used for the treatment of patients with COVID-19, who present signs and symptoms of hyperinflammation. Objectives: To describe the clinical course and behavior of various biomarkers associated with the inflammation and coagulation, in a group of critically ill patients with COVID-19 treated with Jusvinza, compared to a group of patients who did not receive treatment with this peptide. Methods: 40 critically ill patients with COVID-19 were included. The patients were divided into 2 groups: 20 patients were treated with Jusvinza and 20 were not treated with this peptide (control group). Demographic characteristics, comorbidities, vital signs, respiratory parameters and inflammation and coagulation biomarkers were obtained from the medical records of each patient. Results: Treatment with Jusvinza induced a clinical improvement in the patients, associated with the decrease of several inflammation and coagulation biomarkers. Patients treated with Jusvinza had a significantly higher survival than patients not treated with this peptide. Conclusions: Jusvinza is able to control hyperinflammation and hypercoagulation in critical ill patients with COVID-19. © 2021, Editorial Ciencias Medicas. All rights reserved.

4.
Revista Cubana de Pediatria ; 93(3), 2021.
Article in Spanish | Scopus | ID: covidwho-1573353

ABSTRACT

Introduction: The clinical presentation of the disease caused by SARS-CoV-2 is heterogeneous. Patients may be asymptomatic or have mild upper respiratory tract disease, or develop severe pneumonia that can progress to acute respiratory distress syndrome and lead to death. The pathophysiology of severe forms of the disease is characterized by a marked hyperinflammation. Therapies that modulate the immune response can be crucial in treating and preventing this state. The CIGB-258 peptide, brand name Jusvinza, is a therapeutic option for this purpose, due to its immunomodulatory properties. Objective: Describe the clinical evolution of a pediatric patient with severe pneumonia due to SARS-CoV-2, and treated with the CIGB-258 peptide. Case Presentation: A 12-year-old adolescent with refractory epilepsy, infantile cerebral palsy, and an epidemiological history of contact with a patient positive to PCR confirmatory test for SARS-CoV-2 who, eight days after contact, shows clinical, radiographic, and laboratory evidence of severe pneumonia due to COVID-19. Within the therapeutic protocol, he received treatment with CIGB-258 immunomodulatory peptide, with a favorable evolution and hospital discharge. Conclusions: The use of CIGB-258 peptide in the treatment of severe pneumonia due to COVID-19 in pediatrics could contribute to prevent progression to the critical stages of the disease. © 2021, Editorial Ciencias Medicas. All rights reserved.

5.
Cell Stress Chaperones ; 26(3): 515-525, 2021 05.
Article in English | MEDLINE | ID: covidwho-1101014

ABSTRACT

Hyperinflammation distinguishes COVID-19 patients who develop a slight disease or none, from those progressing to severe and critical conditions. CIGB-258 is a therapeutic option for the latter group of patients. This drug is an altered peptide ligand (APL) derived from the cellular stress protein 60 (HSP60). In preclinical models, this peptide developed anti-inflammatory effects and increased regulatory T cell (Treg) activity. Results from a phase I clinical trial with rheumatoid arthritis (RA) patients indicated that CIGB-258 was safe and reduced inflammation. The aim of this study was to examine specific biomarkers associated with hyperinflammation, some cytokines linked to the cytokine storm granzyme B and perforin in a cohort of COVID-19 patients treated with this peptide. All critically ill patients were under invasive mechanical ventilation and received the intravenous administration of 1 or 2 mg of CIGB-258 every 12 h. Seriously ill patients were treated with oxygen therapy receiving 1 mg of CIGB-258 every 12 h and all patients recovered from their severe condition. Biomarker levels associated with hyperinflammation, such as interleukin (IL)-6, IL-10, tumor necrosis factor (TNF-α), granzyme B, and perforin, significantly decreased during treatment. Furthermore, we studied the ability of CIGB-258 to induce Tregs in COVID-19 patients and found that Tregs were induced in all patients studied. Altogether, these results support the therapeutic potential of CIGB-258 for diseases associated with hyperinflammation. Clinical trial registry: RPCEC00000313.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , COVID-19 Drug Treatment , Chaperonin 60/therapeutic use , Cytokine Release Syndrome/drug therapy , Adult , Aged , Aged, 80 and over , Anti-Inflammatory Agents/chemistry , COVID-19/blood , COVID-19/complications , Chaperonin 60/chemistry , Cytokine Release Syndrome/blood , Cytokine Release Syndrome/complications , Female , Humans , Inflammation/blood , Inflammation/complications , Inflammation/drug therapy , Interleukin-10/blood , Interleukin-6/blood , Male , Middle Aged , SARS-CoV-2/drug effects , T-Lymphocytes, Regulatory/drug effects , Tumor Necrosis Factor-alpha/blood , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL