Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Molecules ; 27(22)2022 Nov 21.
Article in English | MEDLINE | ID: covidwho-2123759

ABSTRACT

This work identifies new ligands of the nucleoprotein N of SARS-CoV-2 by in silico screening, which used a new model of N, built from an Alphafold model refined by molecular dynamic simulations. The ligands were neuropeptides, such as substance P (1-7) and enkephalin, bound at a large site of the C-terminal or associated with the N-terminal ß-sheet. The BA4 and BA5 Omicron variants of N also exhibited a large site as in wt N, and an increased flexibility of the BA5 variant, enabling substance P binding. The binding sites of some ligands deduced from modeling in wt N were assessed by mutation studies in surface plasmon resonance experiments. Dynamic light scattering showed that the ligands impeded RNA binding to N, which likely inhibited replication. We suggest that the physiological role of these neuropeptides in neurotransmission, pain and vasodilation for cholecystokinin and substance P could be altered by binding to N. We speculate that N may link between viral replication and multiple pathways leading to long COVID-19 symptoms. Therefore, N may constitute a "danger hub" that needs to be inhibited, even at high cost for the host. Antivirals targeted to N may therefore reduce the risk of brain fog and stroke, and improve patients' health.


Subject(s)
COVID-19 , Neuropeptides , Humans , Nucleoproteins , SARS-CoV-2 , Ligands , Substance P , Synaptic Transmission , Inflammation , Post-Acute COVID-19 Syndrome
2.
Viruses ; 14(10)2022 09 26.
Article in English | MEDLINE | ID: covidwho-2043990

ABSTRACT

SARS-CoV-2 is the etiological agent of COVID-19, an extremely heterogenous disease that can cause severe respiratory failure and critical illness. To date, reliable biomarkers allowing for early patient stratification according to disease severity are still lacking. Calcitonin gene-related peptide (CGRP) is a vasoactive neuropeptide involved in lung pathophysiology and immune modulation and is poorly investigated in the COVID-19 context. In this observational, prospective cohort study, we investigated the correlation between CGRP and clinical disease evolution in hospitalized moderate to severe COVID-19 patients. Between January and May 2021 (Italian third pandemic wave), 135 consecutive SARS-CoV-2 patients were diagnosed as being eligible for the study. Plasma CGRP level evaluation and routine laboratory tests were performed on blood samples collected at baseline and after 7 days of hospitalization. At baseline, the majority our patients had a moderate to severe clinical presentation, and higher plasma CGRP levels predicted a higher risk of in-hospital negative evolution (odds-ratio OR 2.84 [IQR 1.07-7.51]) and were correlated with pulmonary intravascular coagulopathy (OR 2.92 [IQR 1.19-7.17]). Finally, plasma CGRP levels were also correlated with plasma IP10 levels. Our data support a possible crosstalk between the lung and the neuroimmune axis, highlighting a crucial role for plasma CGRP in sustaining COVID-19-related hyperinflammation.


Subject(s)
COVID-19 , Neuropeptides , Humans , Calcitonin Gene-Related Peptide , SARS-CoV-2 , Prospective Studies , Chemokine CXCL10 , Prognosis , Biomarkers
3.
Horm Mol Biol Clin Investig ; 43(3): 363-371, 2022 Sep 01.
Article in English | MEDLINE | ID: covidwho-1846973

ABSTRACT

OBJECTIVES: In this hypothesis paper we explore the underlying mechanisms for long-COVID and how the oxytocinergic neurones could be infected by SARS-CoV-2 leading to a reduction in plasma oxytocin (OXT). Furthermore, we aim to review the relevance of OXT and hypothalamic function in recovery from long-COVID symptoms and pathology, through exploring the pro-health effects of the OXT neuropeptide. METHODS: A review of published literature was surveyed using Google Scholar and PubMed. RESULTS: Numerous experimental data can be shown to correlate with OXT and long-COVID symptoms and conditions, thus providing strong circumstantial evidence to support our hypothesis. It is postulated that the reduction in plasma OXT due to acute and post-viral damage to the hypothalamus and oxytocinergic neurones contributes to the variable multi-system, remitting and relapsing nature of long-COVID. The intranasal route of OXT application was determined to be most appropriate and clinically relevant for the restoration of oxytocinergic function post COVID-19 infection. CONCLUSIONS: We believe it is imperative to further investigate whether OXT alleviates the prolonged suffering of patients with long-COVID. Succinctly, OXT may be the much-needed post-pandemic panacea.


Subject(s)
COVID-19 , Neuropeptides , COVID-19/complications , Humans , Oxytocin/pharmacology , SARS-CoV-2 , Post-Acute COVID-19 Syndrome
4.
6.
Nat Rev Immunol ; 20(10): 592-593, 2020 10.
Article in English | MEDLINE | ID: covidwho-1428862
7.
Cells ; 10(9)2021 09 09.
Article in English | MEDLINE | ID: covidwho-1408628

ABSTRACT

The present study sought to identify gene networks that are hallmarks of the developing inguinal subcutaneous adipose tissue (iWAT) and the interscapular brown adipose tissue (BAT) in the mouse. RNA profiling revealed that the iWAT of postnatal (P) day 6 mice expressed thermogenic and lipid catabolism transcripts, along with the abundance of transcripts associated with the beige adipogenesis program. This was an unexpected finding, as thermogenic BAT was believed to be the only site of nonshivering thermogenesis in the young mouse. However, the transcriptional landscape of BAT in P6 mice suggests that it is still undergoing differentiation and maturation, and that the iWAT temporally adopts thermogenic and lipolytic potential. Moreover, P6 iWAT and adult (P56) BAT were similar in their expression of immune gene networks, but P6 iWAT was unique in the abundant expression of antimicrobial proteins and virus entry factors, including a possible receptor for SARS-CoV-2. In summary, postnatal iWAT development is associated with a metabolic shift from thermogenesis and lipolysis towards fat storage. However, transcripts of beige-inducing signal pathways including ß-adrenergic receptors and interleukin-4 signaling were underrepresented in young iWAT, suggesting that the signals for thermogenic fat differentiation may be different in early postnatal life and in adulthood.


Subject(s)
Adipocytes, Beige/metabolism , Transcription, Genetic , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Animals, Newborn , Biomarkers/metabolism , Cell Cycle/genetics , Gene Expression Regulation, Developmental , Gene Ontology , Gene Regulatory Networks , Male , Mice, Inbred C57BL , Models, Biological , Muscle Development/genetics , Neuropeptides/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
8.
Br J Pharmacol ; 178(21): 4368-4388, 2021 11.
Article in English | MEDLINE | ID: covidwho-1301461

ABSTRACT

BACKGROUND AND PURPOSE: Acute lung injury (ALI), acute respiratory distress syndrome (ARDS) and pulmonary fibrosis remain major causes of morbidity, mortality and a healthcare burden in critically ill patient. There is an urgent need to identify factors causing susceptibility and for the design of new therapeutic agents. Here, we evaluate the effectiveness of the immunomodulatory neuropeptide cortistatin to regulate pulmonary inflammation and fibrosis in vivo. EXPERIMENTAL APPROACH: ALI/ARDS and pulmonary fibrosis were induced experimentally in wild-type and cortistatin-deficient mice by pulmonary infusion of the bacterial endotoxin LPS or the chemotherapeutic drug bleomycin, and the histopathological signs, pulmonary leukocyte infiltration and cytokines, and fibrotic markers were evaluated. KEY RESULTS: Partially deficient mice in cortistatin showed exacerbated pulmonary damage, pulmonary inflammation, alveolar oedema and fibrosis, and subsequent increased respiratory failure and mortality when challenged to LPS or bleomycin, even at low doses. Treatment with cortistatin reversed these aggravated phenotypes and protected from progression to severe ARDS and fibrosis, after high exposure to both injury agents. Moreover, cortistatin-deficient pulmonary macrophages and fibroblasts showed exaggerated ex vivo inflammatory and fibrotic responses, and treatment with cortistatin impaired their activation. Finally, the protective effects of cortistatin in ALI and pulmonary fibrosis were partially inhibited by specific antagonists for somatostatin and ghrelin receptors. CONCLUSION AND IMPLICATIONS: We identified cortistatin as an endogenous inhibitor of pulmonary inflammation and fibrosis. Deficiency in cortistatin could be a marker of poor prognosis in inflammatory/fibrotic pulmonary disorders. Cortistatin-based therapies could emerge as attractive candidates to treat severe ALI/ARDS, including SARS-CoV-2-associated ARDS.


Subject(s)
Inflammation , Neuropeptides , Pneumonia , Animals , Disease Models, Animal , Fibrosis , Inflammation/drug therapy , Inflammation/pathology , Lipopolysaccharides , Lung/pathology , Mice , Neuropeptides/pharmacology , Pneumonia/chemically induced , Pneumonia/drug therapy
9.
Neuropeptides ; 89: 102159, 2021 Oct.
Article in English | MEDLINE | ID: covidwho-1225350

ABSTRACT

T cells of aged people, and of patients with either cancer or severe infections (including COVID-19), are often exhausted, senescent and dysfunctional, leading to increased susceptibilities, complications and mortality. Neurotransmitters and Neuropeptides bind their receptors in T cells, and induce multiple beneficial T cell functions. Yet, T cells of different people vary in the expression levels of Neurotransmitter and Neuropeptide receptors, and in the magnitude of the corresponding effects. Therefore, we performed an individual-based study on T cells of 3 healthy subjects, and 3 Hepatocellular Carcinoma (HCC) patients. HCC usually develops due to chronic inflammation. The inflamed liver induces reduction and inhibition of CD4+ T cells and Natural Killer (NK) cells. Immune-based therapies for HCC are urgently needed. We tested if selected Neurotransmitters and Neuropeptides decrease the key checkpoint protein PD-1 in human T cells, and increase proliferation and killing of HCC cells. First, we confirmed human T cells express all dopamine receptors (DRs), and glutamate receptors (GluRs): AMPA-GluR3, NMDA-R and mGluR. Second, we discovered that either Dopamine, Glutamate, GnRH-II, Neuropeptide Y and/or CGRP (10nM), as well as DR and GluR agonists, induced the following effects: 1. Decreased significantly both %PD-1+ T cells and PD-1 expression level per cell (up to 60% decrease, within 1 h only); 2. Increased significantly the number of T cells that proliferated in the presence of HCC cells (up to 7 fold increase), 3. Increased significantly T cell killing of HCC cells (up to 2 fold increase). 4. Few non-conventional combinations of Neurotransmitters and Neuropeptides had surprising synergistic beneficial effects. We conclude that Dopamine, Glutamate, GnRH-II, Neuropeptide Y and CGRP, alone or in combinations, can decrease % PD-1+ T cells and PD-1 expression per cell, in T cells of both healthy subjects and HCC patients, and increase their proliferation in response to HCC cells and killing of HCC cells. Yet, testing T cells of many more cancer patients is absolutely needed. Based on these findings and previous ones, we designed a novel "Personalized Adoptive Neuro-Immunotherapy", calling for validation of safety and efficacy in clinical trials.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Proliferation/drug effects , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Neuropeptides/pharmacology , Neurotransmitter Agents/pharmacology , Programmed Cell Death 1 Receptor/biosynthesis , Programmed Cell Death 1 Receptor/genetics , T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/metabolism , COVID-19/complications , Carcinoma, Hepatocellular/pathology , Dopamine/pharmacology , Dopamine Agonists/pharmacology , Humans , Immunotherapy , Killer Cells, Natural/metabolism , Liver Neoplasms/pathology , Receptors, Glutamate/drug effects , Receptors, Neuropeptide/metabolism , Receptors, Neurotransmitter/metabolism
10.
Redox Biol ; 38: 101794, 2021 01.
Article in English | MEDLINE | ID: covidwho-922123

ABSTRACT

Acute lung injury (ALI), a devastating illness induced by systemic inflammation e.g., sepsis or local lung inflammation e.g., COVID-19 mediated severe pneumonia, has an unacceptably high mortality and has no effective therapy. ALI is associated with increased pulmonary microvascular hyperpermeability and alveolar flooding. The small Rho GTPases, RhoA and Rac1 are central regulators of vascular permeability through cytoskeleton rearrangements. RhoA and Rac1 have opposing functional outcome: RhoA induces an endothelial contractile phenotype and barrier disruption, while Rac1 stabilizes endothelial junctions and increases barrier integrity. In ALI, RhoA activity is increased while Rac1 activity is reduced. We have shown that the activation of RhoA in lipopolysaccharide (LPS)-mediated ALI, is dependent, at least in part, on a single nitration event at tyrosine (Y)34. Thus, the purpose of this study was to determine if the inhibition of Rac1 is also dependent on its nitration. Our data show that Rac1 inhibition by LPS is associated with its nitration that mass spectrometry identified as Y32, within the switch I region adjacent to the nucleotide-binding site. Using a molecular modeling approach, we designed a nitration shielding peptide for Rac1, designated NipR2 (nitration inhibitor peptide for the Rho GTPases 2), which attenuated the LPS-induced nitration of Rac1 at Y32, preserves Rac1 activity and attenuates the LPS-mediated disruption of the endothelial barrier in human lung microvascular endothelial cells (HLMVEC). Using a murine model of ALI induced by intratracheal installation of LPS we found that NipR2 successfully prevented Rac1 nitration and Rac1 inhibition, and more importantly attenuated pulmonary inflammation, reduced lung injury and prevented the loss of lung function. Together, our data identify a new post-translational mechanism of Rac1 inhibition through its nitration at Y32. As NipR2 also reduces sepsis induced ALI in the mouse lung, we conclude that Rac1 nitration is a therapeutic target in ALI.


Subject(s)
Acute Lung Injury , Blood-Air Barrier , COVID-19 , Endothelial Cells , Lipopolysaccharides/toxicity , Neuropeptides/metabolism , SARS-CoV-2/metabolism , rac1 GTP-Binding Protein/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/enzymology , Acute Lung Injury/pathology , Acute Lung Injury/virology , Animals , Blood-Air Barrier/enzymology , Blood-Air Barrier/pathology , Blood-Air Barrier/virology , COVID-19/chemically induced , COVID-19/enzymology , COVID-19/pathology , Cell Line , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Cells/virology , Humans , Male , Mice , Neuropeptides/genetics , rac1 GTP-Binding Protein/genetics
12.
Inflamm Bowel Dis ; 26(8): e89-e91, 2020 07 17.
Article in English | MEDLINE | ID: covidwho-343390

ABSTRACT

Severe Acute Respiratory Syndrome Coronavirus-2 (SAR-CoV-2) has been shown to invade brain tissue. Based on the evolutionary similarity with SARS-CoV, researchers propose that SARS-CoV-2 can invade the olfactory bulb and gastrointestinal (GI) system through angiotensin-converting enzyme 2. However, how SARS-CoV-2 causes neurological or GI symptoms is not clear. Many suggested intestinal and neural inflammations, caused by viral invasion, as the most likely reason for the GI and neurological symptoms; however, the patients with coronavirus disease 2019 (COVID-19) without neurological or GI symptoms indicate that this is not the case. The gut-brain axis could explain the reason for why some with COVID-19 do not have these symptoms. COVID-19 patients mostly show respiratory distress first, then diarrhea, anorexia, stroke, or loss of consciousness comes into view. Obviously, GI invasion is a mechanical process that begins with oral invasion and, therefore, most probably exists before the brain invasion, as indicated in case reports. However, when the GI tract is invaded, the virus may enter the central nervous system through vascular and lymphatic systems or the vagal nerve. SARS-CoV-2 can infect leukocytes and migrate with them into the brain, or the viral particles can be directly transported across the blood-brain barrier to the brain. Also, more recent research has revealed that SARS-CoV-2 can invade the peripheral lymphatic vessels connecting with the glymphatic system of the brain. The temporal correlation between neurological and gastrointestinal symptoms suggests the lymph vessels around the GI tract, the vascular system, or the gut-brain axis (enteric nervous system) as the most likely entry route for SARS-CoV-2 to the brain.


Subject(s)
Coronavirus Infections/epidemiology , Enteric Nervous System/physiopathology , Gastrointestinal Diseases/epidemiology , Nervous System Diseases/epidemiology , Pandemics/statistics & numerical data , Pneumonia, Viral/epidemiology , Severe Acute Respiratory Syndrome/epidemiology , COVID-19 , Comorbidity , Coronavirus Infections/diagnosis , Coronavirus Infections/therapy , Female , Gastrointestinal Diseases/diagnosis , Gastrointestinal Diseases/microbiology , Global Health , Humans , Male , Nervous System Diseases/diagnosis , Nervous System Diseases/therapy , Neuropeptides , Oligopeptides , Pneumonia, Viral/diagnosis , Pneumonia, Viral/therapy , Prevalence , Risk Assessment , Severe Acute Respiratory Syndrome/diagnosis , Severe Acute Respiratory Syndrome/therapy , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL