Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Front Biosci (Landmark Ed) ; 26(10): 948-961, 2021 10 30.
Article in English | MEDLINE | ID: covidwho-1498509

ABSTRACT

Background: Corona Virus Disease 2019 (COVID-19) is an acute respiratory infectious disease caused by severe respiratory syndrome coronavirus 2 (SARS-CoV-2). The primary pathogenesis is over-activation of the immune system. SARS-CoV-2 continues to mutate and spread rapidly and no effective treatment options are yet available. Mesenchymal stem cells (MSCs) are known to induce anti-inflammatory macrophages, regulatory T cells and dendritic cells. There are a rapidly increasing number of clinical investigations of cell-based therapy approaches for COVID-19. Objective: To summarize the pathogenic mechanism of SARS-CoV-2, and systematically formulated the immunomodulation of COVID-19 by MSCs and their exosomes, as well as research progress. Method: Searching PubMed, clinicaltrials.gov and Chictr.cn for eligible studies to be published or registered by May 2021. Main keywords and search strategies were as follows: ((Mesenchymal stem cells) OR (MSCs)) AND (COVID-19). Results: MSCs regulate the immune system to prevent cytokine release syndrome (CRS) and to promote endogenous repair by releasing various paracrine factors and exosomes. Conclusions: MSC therapy is thus a promising candidate for COVID-19.


Subject(s)
COVID-19/therapy , Exosomes/transplantation , Immunomodulation/immunology , Lung Injury/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , COVID-19/epidemiology , COVID-19/virology , Clinical Trials as Topic , Exosomes/immunology , Exosomes/metabolism , Humans , Lung Injury/physiopathology , Lung Injury/virology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Outcome Assessment, Health Care/methods , Outcome Assessment, Health Care/statistics & numerical data , Pandemics , Regeneration/immunology , Regeneration/physiology , SARS-CoV-2/immunology , SARS-CoV-2/physiology
2.
Br J Anaesth ; 127(4): 648-659, 2021 Oct.
Article in English | MEDLINE | ID: covidwho-1329691

ABSTRACT

Mechanical ventilation induces a number of systemic responses for which the brain plays an essential role. During the last decade, substantial evidence has emerged showing that the brain modifies pulmonary responses to physical and biological stimuli by various mechanisms, including the modulation of neuroinflammatory reflexes and the onset of abnormal breathing patterns. Afferent signals and circulating factors from injured peripheral tissues, including the lung, can induce neuronal reprogramming, potentially contributing to neurocognitive dysfunction and psychological alterations seen in critically ill patients. These impairments are ubiquitous in the presence of positive pressure ventilation. This narrative review summarises current evidence of lung-brain crosstalk in patients receiving mechanical ventilation and describes the clinical implications of this crosstalk. Further, it proposes directions for future research ranging from identifying mechanisms of multiorgan failure to mitigating long-term sequelae after critical illness.


Subject(s)
Brain/metabolism , Lung Injury/physiopathology , Respiration, Artificial/methods , Animals , Central Nervous System/metabolism , Critical Illness , Humans , Multiple Organ Failure/physiopathology , Positive-Pressure Respiration/methods
3.
Physiol Rep ; 9(13): e14802, 2021 07.
Article in English | MEDLINE | ID: covidwho-1305905

ABSTRACT

In severe acute respiratory distress syndrome (ARDS), extracorporeal membrane oxygenation (ECMO) is a life-prolonging treatment, especially among COVID-19 patients. Evaluation of lung injury progression is challenging with current techniques. Diagnostic imaging or invasive diagnostics are risky given the difficulties of intra-hospital transportation, contraindication of biopsies, and the potential for the spread of infections, such as in COVID-19 patients. We have recently shown that particle flow rate (PFR) from exhaled breath could be a noninvasive, early detection method for ARDS during mechanical ventilation. We hypothesized that PFR could also measure the progress of lung injury during ECMO treatment. Lipopolysaccharide (LPS) was thus used to induce ARDS in pigs under mechanical ventilation. Eight were connected to ECMO, whereas seven animals were not. In addition, six animals received sham treatment with saline. Four human patients with ECMO and ARDS were also monitored. In the pigs, as lung injury ensued, the PFR dramatically increased and a particular spike followed the establishment of ECMO in the LPS-treated animals. PFR remained elevated in all animals with no signs of lung recovery. In the human patients, in the two that recovered, PFR decreased. In the two whose lung function deteriorated while on ECMO, there was increased PFR with no sign of recovery in lung function. The present results indicate that real-time monitoring of PFR may be a new, complementary approach in the clinic for measurement of the extent of lung injury and recovery over time in ECMO patients with ARDS.


Subject(s)
COVID-19/physiopathology , Lipopolysaccharides/toxicity , Lung Injury/physiopathology , Lung/physiopathology , Particulate Matter/analysis , Respiratory Distress Syndrome/physiopathology , Animals , Blood Gas Analysis/methods , COVID-19/chemically induced , Extracorporeal Membrane Oxygenation/methods , Lung/drug effects , Lung Injury/chemically induced , Particulate Matter/adverse effects , Respiration, Artificial/methods , Respiratory Distress Syndrome/chemically induced , Swine
4.
Crit Care ; 25(1): 74, 2021 02 19.
Article in English | MEDLINE | ID: covidwho-1090628

ABSTRACT

BACKGROUND: Biomarkers can be used to detect the presence of endothelial and/or alveolar epithelial injuries in case of ARDS. Angiopoietin-2 (Ang-2), soluble intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion protein-1 (VCAM-1), P-selectin and E-selectin are biomarkers of endothelial injury, whereas the receptor for advanced glycation end-products (RAGE) reflects alveolar epithelial injury. The aims of this study were to evaluate whether the plasma concentration of the above-mentioned biomarkers was different 1) in survivors and non-survivors of COVID-19-related ARDS and 2) in COVID-19-related and classical ARDS. METHODS: This prospective study was performed in two COVID-19-dedicated Intensive Care Units (ICU) and one non-COVID-19 ICU at Ferrara University Hospital. A cohort of 31 mechanically ventilated patients with COVID-19 ARDS and a cohort of 11 patients with classical ARDS were enrolled. Ang-2, ICAM-1, VCAM-1, P-selectin, E-selectin and RAGE were determined with a bead-based multiplex immunoassay at three time points: inclusion in the study (T1), after 7 ± 2 days (T2) and 14 ± 2 days (T3). The primary outcome was to evaluate the plasma trend of the biomarker levels in survivors and non-survivors. The secondary outcome was to evaluate the differences in respiratory mechanics variables and gas exchanges between survivors and non-survivors. Furthermore, we compared the plasma levels of the biomarkers at T1 in patients with COVID-19-related ARDS and classical ARDS. RESULTS: In COVID-19-related ARDS, the plasma levels of Ang-2 and ICAM-1 at T1 were statistically higher in non-survivors than survivors, (p = 0.04 and p = 0.03, respectively), whereas those of P-selectin, E-selectin and RAGE did not differ. Ang-2 and ICAM-1 at T1 were predictors of mortality (AUROC 0.650 and 0.717, respectively). At T1, RAGE and P-selectin levels were higher in classical ARDS than in COVID-19-related ARDS. Ang-2, ICAM-1 and E-selectin were lower in classical ARDS than in COVID-19-related ARDS (all p < 0.001). CONCLUSIONS: COVID-19 ARDS is characterized by an early pulmonary endothelial injury, as detected by Ang-2 and ICAM-1. COVID-19 ARDS and classical ARDS exhibited a different expression of biomarkers, suggesting different pathological pathways. Trial registration NCT04343053 , Date of registration: April 13, 2020.


Subject(s)
Biomarkers/analysis , Lung Injury/diagnosis , Respiration, Artificial/adverse effects , Aged , Antigens, Neoplasm/analysis , Antigens, Neoplasm/blood , Area Under Curve , COVID-19/blood , COVID-19/prevention & control , Cohort Studies , E-Selectin/analysis , E-Selectin/blood , Female , Humans , Intensive Care Units/organization & administration , Intensive Care Units/statistics & numerical data , Intercellular Adhesion Molecule-1/analysis , Intercellular Adhesion Molecule-1/blood , Lung Injury/blood , Lung Injury/physiopathology , Male , Middle Aged , Mitogen-Activated Protein Kinases/analysis , Mitogen-Activated Protein Kinases/blood , P-Selectin/analysis , P-Selectin/blood , Prospective Studies , ROC Curve , Respiration, Artificial/standards , Respiration, Artificial/statistics & numerical data , Respiratory Distress Syndrome/blood , Respiratory Distress Syndrome/physiopathology , Versicans/analysis , Versicans/blood , Vesicular Transport Proteins/analysis , Vesicular Transport Proteins/blood
5.
Open Heart ; 7(2)2020 12.
Article in English | MEDLINE | ID: covidwho-1066930

ABSTRACT

SARS-CoV-2 is the virus responsible for the ongoing COVID-19 outbreak. The virus uses ACE2 receptor for viral entry. ACE2 is part of the counter-regulatory renin-angiotensin-aldosterone system and is also expressed in the lower respiratory tract along the alveolar epithelium. There is, however, significant controversy regarding the role of ACE2 expression in COVID-19 pathogenesis. Some have argued that decreasing ACE2 expression would result in decreased susceptibility to the virus by decreasing available binding sites for SARS-CoV-2 and restricting viral entry into the cells. Others have argued that, like the pathogenesis of other viral pneumonias, including those stemming from previous severe acute respiratory syndrome (SARS) viruses, once SARS-CoV-2 binds to ACE2, it downregulates ACE2 expression. Lack of the favourable effects of ACE2 might exaggerate lung injury by a variety of mechanisms. In order to help address this controversy, we conducted a literature search and review of relevant preclinical and clinical publications pertaining to SARS-CoV-2, COVID-19, ACE2, viral pneumonia, SARS, acute respiratory distress syndrome and lung injury. Our review suggests, although controversial, that patients at increased susceptibility to COVID-19 complications may have reduced baseline ACE2, and by modulating ACE2 expression one can possibly improve COVID-19 outcomes. Herein, we elucidate why and how this potential mechanism might work.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/diagnosis , COVID-19/metabolism , Renin-Angiotensin System/drug effects , SARS-CoV-2/genetics , Adult , Angiotensin Receptor Antagonists/pharmacology , Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , COVID-19/virology , Down-Regulation , Female , Humans , Immunity/immunology , Lung Injury/drug therapy , Lung Injury/physiopathology , Male , Mice , Middle Aged , Models, Animal , Pneumonia, Viral/drug therapy , Respiratory Distress Syndrome/drug therapy , Risk Factors , SARS-CoV-2/drug effects , Virus Internalization/drug effects , COVID-19 Drug Treatment
6.
J Med Virol ; 93(4): 2505-2512, 2021 04.
Article in English | MEDLINE | ID: covidwho-1023298

ABSTRACT

To investigate the dynamic changes of Krebs von den Lungen-6 (KL-6) among patients with coronavirus disease 2019 (COVID-19) and the role of KL-6 as a noninvasive biomarker for predicting long-term lung injury, the clinical information and laboratory tests of 166 COVID-19 patients were collected, and a correlation analysis between KL-6 and other parameters was conducted. There were 17 (10.2%, 17/166) severe/critical and 149 (89.8%, 149/166) mild COVID-19 patients in our cohort. Serum KL-6 was significantly higher in severe/critical COVID-19 patients than in mild patients (median 898.0 vs. 451.2 U/ml, p < .001). KL-6 was next confirmed to be a sensitive and specific biomarker for distinguishing mild and severe/critical patients and correlate to computed tomography lung lesions areas. Serum KL-6 concentration during the follow-up period (>100 days postonset) was well correlated to those concentrations within 10 days postonset (Pearson r = .867, p < .001), indicating the prognostic value of KL-6 levels in predicting lung injury after discharge. Finally, elevated KL-6 was found to be significantly correlated to coagulation disorders, and T cells subsets dysfunctions. In summary, serum KL-6 is a biomarker for assessing COVID-19 severity and predicting the prognosis of lung injury of discharged patients.


Subject(s)
COVID-19/blood , Lung Injury/blood , Mucin-1/blood , Adult , Aged , Biomarkers/blood , COVID-19/diagnostic imaging , Female , Humans , Lung/diagnostic imaging , Lung/physiopathology , Lung Injury/diagnostic imaging , Lung Injury/physiopathology , Male , Middle Aged , Prognosis , Retrospective Studies , SARS-CoV-2/isolation & purification , Severity of Illness Index , Tomography, X-Ray Computed/methods
7.
Int J Mol Sci ; 21(18)2020 Sep 17.
Article in English | MEDLINE | ID: covidwho-918204

ABSTRACT

Acute respiratory distress syndrome (ARDS) is associated with high morbidity and mortality, and current management has a dramatic impact on healthcare resource utilization. While our understanding of this disease has improved, the majority of treatment strategies remain supportive in nature and are associated with continued poor outcomes. There is a dramatic need for the development and breakthrough of new methods for the treatment of ARDS. Isolated machine lung perfusion is a promising surgical platform that has been associated with the rehabilitation of injured lungs and the induction of molecular and cellular changes in the lung, including upregulation of anti-inflammatory and regenerative pathways. Initially implemented in an ex vivo fashion to evaluate marginal donor lungs prior to transplantation, recent investigations of isolated lung perfusion have shifted in vivo and are focused on the management of ARDS. This review presents current tenants of ARDS management and isolated lung perfusion, with a focus on how ex vivo lung perfusion (EVLP) has paved the way for current investigations utilizing in vivo lung perfusion (IVLP) in the treatment of severe ARDS.


Subject(s)
Inflammation/therapy , Lung Injury/therapy , Perfusion/methods , Respiratory Distress Syndrome/therapy , Animals , History, 19th Century , History, 20th Century , History, 21st Century , Humans , Inflammation/physiopathology , Lung Injury/physiopathology , Perfusion/history , Perfusion/instrumentation , Respiratory Distress Syndrome/diagnostic imaging , Tissue Donors
8.
Ann Am Thorac Soc ; 17(8): 918-921, 2020 08.
Article in English | MEDLINE | ID: covidwho-853546

ABSTRACT

Amid efforts to care for the large number of patients with coronavirus disease (COVID-19), there has been considerable speculation about whether the lung injury seen in these patients is different than acute respiratory distress syndrome from other causes. One idea that has garnered considerable attention, particularly on social media and in free open-access medicine, is the notion that lung injury due to COVID-19 is more similar to high-altitude pulmonary edema (HAPE). Drawing on this concept, it has also been proposed that treatments typically employed in the management of HAPE and other forms of acute altitude illness-pulmonary vasodilators and acetazolamide-should be considered for COVID-19. Despite some similarities in clinical features between the two entities, such as hypoxemia, radiographic opacities, and altered lung compliance, the pathophysiological mechanisms of HAPE and lung injury due to COVID-19 are fundamentally different, and the entities cannot be viewed as equivalent. Although of high utility in the management of HAPE and acute mountain sickness, systemically delivered pulmonary vasodilators and acetazolamide should not be used in the treatment of COVID-19, as they carry the risk of multiple adverse consequences, including worsened ventilation-perfusion matching, impaired carbon dioxide transport, systemic hypotension, and increased work of breathing.


Subject(s)
Altitude Sickness , Coronavirus Infections , Hypertension, Pulmonary , Pandemics , Pneumonia, Viral , Respiratory Distress Syndrome , Acetazolamide/pharmacology , Altitude Sickness/physiopathology , Altitude Sickness/therapy , Betacoronavirus/isolation & purification , COVID-19 , Carbonic Anhydrase Inhibitors/pharmacology , Coronavirus Infections/complications , Coronavirus Infections/drug therapy , Coronavirus Infections/physiopathology , Coronavirus Infections/therapy , Humans , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/therapy , Lung Injury/etiology , Lung Injury/physiopathology , Lung Injury/therapy , Nifedipine/pharmacology , Pneumonia, Viral/physiopathology , Pneumonia, Viral/therapy , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/physiopathology , Respiratory Distress Syndrome/therapy , SARS-CoV-2 , Vasodilator Agents/pharmacology , COVID-19 Drug Treatment
9.
Crit Care ; 24(1): 494, 2020 08 10.
Article in English | MEDLINE | ID: covidwho-704904

ABSTRACT

Deterioration of lung function during the first week of COVID-19 has been observed when patients remain with insufficient respiratory support. Patient self-inflicted lung injury (P-SILI) is theorized as the responsible, but there is not robust experimental and clinical data to support it. Given the limited understanding of P-SILI, we describe the physiological basis of P-SILI and we show experimental data to comprehend the role of regional strain and heterogeneity in lung injury due to increased work of breathing.In addition, we discuss the current approach to respiratory support for COVID-19 under this point of view.


Subject(s)
Coronavirus Infections/physiopathology , Disease Progression , Lung Injury/physiopathology , Pneumonia, Viral/physiopathology , Work of Breathing/physiology , COVID-19 , Coronavirus Infections/therapy , Critical Care , Humans , Lung Injury/etiology , Pandemics , Pneumonia, Viral/therapy , Randomized Controlled Trials as Topic , Respiration, Artificial
11.
ACS Chem Neurosci ; 11(15): 2156-2158, 2020 08 05.
Article in English | MEDLINE | ID: covidwho-679395

ABSTRACT

Lung injury with COVID-19 may be due to a complex underlying pathophysiology. Cytokine release syndrome appears to be a catalyst of different inflammatory pathways promoting lung parenchymal injury and thromboembolic phenomena ("dual hit" injury). Recently, severe neurological manifestations such as acute disseminated encephalomyelitis, which may be not linked to lung pathology, have been identified in COVID-19, contributing thus further to the versatility of its clinical features.


Subject(s)
Betacoronavirus/metabolism , Coronavirus Infections/metabolism , Cytokines/metabolism , Lung Injury/metabolism , Pneumonia, Viral/metabolism , Animals , COVID-19 , Coronavirus Infections/complications , Coronavirus Infections/physiopathology , Humans , Lung Injury/etiology , Lung Injury/physiopathology , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/physiopathology , SARS-CoV-2
12.
Cells ; 9(6)2020 06 10.
Article in English | MEDLINE | ID: covidwho-592028

ABSTRACT

Pulmonary failure is the main cause of morbidity and mortality in the human chromosomal instability syndrome Ataxia-telangiectasia (A-T). Major phenotypes include recurrent respiratory tract infections and bronchiectasis, aspiration, respiratory muscle abnormalities, interstitial lung disease, and pulmonary fibrosis. At present, no effective pulmonary therapy for A-T exists. Cell therapy using adipose-derived mesenchymal stromal/stem cells (ASCs) might be a promising approach for tissue regeneration. The aim of the present project was to investigate whether ASCs migrate into the injured lung parenchyma of Atm-deficient mice as an indication of incipient tissue damage during A-T. Therefore, ASCs isolated from luciferase transgenic mice (mASCs) were intravenously transplanted into Atm-deficient and wild-type mice. Retention kinetics of the cells were monitored using in vivo bioluminescence imaging (BLI) and completed by subsequent verification using quantitative real-time polymerase chain reaction (qRT-PCR). The in vivo imaging and the qPCR results demonstrated migration accompanied by a significantly longer retention time of transplanted mASCs in the lung parenchyma of Atm-deficient mice compared to wild type mice. In conclusion, our study suggests incipient damage in the lung parenchyma of Atm-deficient mice. In addition, our data further demonstrate that a combination of luciferase-based PCR together with BLI is a pivotal tool for tracking mASCs after transplantation in models of inflammatory lung diseases such as A-T.


Subject(s)
Ataxia Telangiectasia/complications , Lung Diseases/etiology , Lung Injury/etiology , Mesenchymal Stem Cells/metabolism , Animals , Ataxia Telangiectasia/pathology , Disease Models, Animal , Humans , Lung Diseases/physiopathology , Lung Injury/physiopathology , Mice , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL