hmgb1, microrna, inflammation, and coagulation charles lewis, … · 2020. 5. 12. · nlrp3 is a...

23
HMGB1, microRNA, Inflammation, and Coagulation Charles Lewis, MD MPH NOTICE: This is a largely theoretical discussion of agents that may be useful in the treatment of COVID-19, sepsis and cytokine storm. It is being posted to suggest potential lines of inquiry for researchers interested in conducting laboratory research or use in well-controlled, sanctioned clinical trials. It should not be assumed to be correct. It should not be construed as medical advice. In COVID-19, there is both inhibition of immune processes and hyperactivity of immune systems. In the simplest terms, the adaptive immune system is down-regulated making the production of protective antibodies inefficient, while parts of the innate immune system are quiescent and other parts go into hyper- drive, causing tissue injury while not effectively stemming viral replication. One part of innate immunity that is upregulated is illustrated below.

Upload: others

Post on 10-Feb-2021

6 views

Category:

Documents


0 download

TRANSCRIPT

  • HMGB1, microRNA, Inflammation, and Coagulation Charles Lewis, MD MPH

    NOTICE: This is a largely theoretical discussion of agents that may be useful in the treatment of COVID-19,

    sepsis and cytokine storm. It is being posted to suggest potential lines of inquiry for researchers interested

    in conducting laboratory research or use in well-controlled, sanctioned clinical trials. It should not be

    assumed to be correct. It should not be construed as medical advice.

    In COVID-19, there is both inhibition of immune processes and hyperactivity of immune systems. In the

    simplest terms, the adaptive immune system is down-regulated making the production of protective

    antibodies inefficient, while parts of the innate immune system are quiescent and other parts go into hyper-

    drive, causing tissue injury while not effectively stemming viral replication.

    One part of innate immunity that is upregulated is illustrated below.

  • ZBP1 is an intracellular protein that is part of the innate immune system. It is activated by the presence of

    viral RNA or DNA. Both ZBP1 and TRIF (not shown here) activate RIPK3. RIPK3 activates the “necrosome”

    which activates numerous functions that result in the death of cells infected by a virus. As part of this

    process, HMGB1 is released from the cell. HMGB1 act locally as a Damage-Associated Molecular Pattern

    (DAMP) signal, promoting a higher level of inflammation. ZBP1 also activates Interferon Related Factors,

    IRF3 and IRF7, which are nuclear transcription factors for the expression of interferon α and β (IFNα and

    IFNβ). ZBP1 also activates JNK. SARS-CoV-2 is a single stranded RNA virus. Thus, presence of SARS-CoV-2

    RNA in the cells activates ZBP1.

    The SARS-CoV viral protein Papain-Like Protease (PLPro) inhibits the production of IFNα and IFNβ. IFNα

    and IFNβ are essential to both innate and adaptive immunity. In adaptive immunity, they stimulate T-

    helper cells which help coordinate B-cell production of antibodies, among other functions. These cytikines

    also stimulate Natural Killer cell function.

    Another SARS-CoV viral protein, ORF3a also activates the NRLP3 inflammasome via TRAF3,1 and ACS.

    This stimulates the assembly of the NRLP3 inflammasome that promotes the pyroptosis form of

    programmed cell death. NLRP3 is a cellular stress sensor. Activation of the NLRP3 inflammasome leads to

    Casp1 activation and IL-1β and IL-18 release and active secretion of HMGB1. In this process HMGB1 is

    released along with inflammatory cytokines. HMGB1 binds with TLR4 and TRL2, stimulating additional

    inflammatory activity.

    This virus may also causes inflammation by depleting ACE2, an anti-inflammatory protein. ACE2 is the

    protein the SARS-CoV-2 binds to on the surface of the alveolar cells and certain other cells of the body.

    Without an adequate anti-viral and innate immune signalling response from the target cells of SARS-CoV-2,

    i.e.; the alveolar endothelial cells, there is delayed protective immune response and poorly control over virus

    proliferation. There is likely little cytokine signaling to myelogenous immune cells, (NK cells, Monocytes, T

    helper cells) until the infected cells start necroptosis or form inflammasomes that induce pyroptosis. Under

    these cascades, there is the late release of IL-1, IL-33 and DAMPSs including HMGB1, ATP and

    mitochondrial DNA.

    Once DAMPS are released, they activate macrophages and monocytes. HMGB1 and IL-1 and other DAMPs

    thus can act as delayed stimuli for immune response. HMGB1 is ligand for the pattern recognition receptors

    (PRRs) TLR4 and TLR2 receptors that promote the inflammatory response in white blood cells. TLR4

    activation by DAMPS activates NK-κB as well as other transcription factors for the inflammatory response

    that produce TNFα and other cytokines.

    DAMPs are largely released as a part of pyroptosis and necroptosis; it can be understood that these

    cytokines signal the need for “a cleanup crew” to breakdown and clear up dead and injured cells after injury.

    This mechanism did not evolve to act as a primary immune mechanism to get rid of a virus, but rather as

    demolition after injury, prior to repair. When this process becomes the primary immune response, it

    functions through tissue destruction. Thus, DAMPs act as cytokines for macrophages to move into the

    alveoli, where they clear up dead cells, but also cause injury. Additionally, filling the alveoli with white blood

    cells, the leaking of serous fluid, and the injury they provoke can cause acute respiratory distress syndrome

    (ARDS). It is neutrophils and macrophages that have migrated to the lungs which are the major source of

    cytokines fueling the inflammatory reaction.2

  • Over-production of TNFα or vigorous releases of HMGB1 fuel the acceleration of the inflammatory

    response. It can become like a fire that will burn until it runs out of fuel. While TNFα and INF appear in

    hours, and can remit as quickly, HMGB1 release from the cell nucleus into the cytoplasm and subsequent

    release from the cell can take one to two days. After that, HMGB1 remains in the blood stream for several

    days, and can affect most of the organs as well as white blood cells. In the lungs it causes neutrophil

    infiltration, edema and alveolar injury.

    In SARS and severe influenza, the development of a sepsis-like lung disease can develop. There is

    recruitment of macrophages to the lung and destruction of the alveolar endothelium. In severe COVID-19

    there are cytokine storm-like processes, however we now understand that the primary disease is vascular,

    involving the endothelium, and the lung disease is at least usually a secondary effect.

    There is emerging evidence that there are two different presentations of severe COVID-19; a wet and a dry

    form.3 Recognizing these two presentations of severe COVID-19 not just important in the respiratory

    management of the disease, but also because it suggests that the two different presentations may involve

    different immune pathways.

    In the dry form, which is more common and less severe, the amount of fluid in the lung is not greatly

    elevated and the lungs have near normal compliance. The patient is able to breathe, but does not get

    sufficient oxygen. It is thought that there may be either vasoconstriction of the pulmonary venules so that

    the blood is not getting sufficiently oxygenated, and/or there may be micro-thrombi, blocking blood flow to

    the pulmonary capillaries. Another additional theory is that the red blood cells are not carrying oxygen

    normally. In the dry form of COVID-19, the lung maintains good compliance (elasticity) and the patient can

    move air in and out of the lungs without excessive difficulty, but is not having good gas exchange in the

    lungs. Intubation with the use of a ventilator with PEEP (positive end expiratory pressure) may not be

    helpful and may be dangerous to the patient; however oxygen therapy is typically required. The dry form

    appears to be the more common, or perhaps may just appear earlier in the disease.

    In the wet form, the lung has a high level of fluid in the alveoli. This presentation is like the typical ARDS

    (acute respiratory distress syndrome) with macrophage and neutrophil infiltration, edema and alveolar

    injury. Here therapy with a ventilator and PEEP to expand the alveoli may be helpful.

  • In COVID and influenza, OxPL (oxidized phospholipids) are released from virally infected cells as a result of

    oxidative stress.4 This response is protective, as it allows the mounting of the TLR4 (Toll-like receptor-4)

    triggered immune defense.5 TLR4 is a cell surface protein that is activated by DAMPS and PAMPs

    (pathogen-associated molecular patterns). Several Toll-like Receptors activate MyD88. MyD88 mediates

    several downstream events culminating in the release of NF-κB, a transcription factor for TNFα, IL-1β, IL6,

    and IL-12. MyD88 also promotes the transcription for INFα and INFβ.

    As a secondary trigger, intracellular TLR3 (Toll-like receptor-3) response is activated by the presence of

    single-stranded RNA (ssRNA) in the cell. TLR3 triggers TRIF, which is a MyD88-independent pathway that

    also promotes INF-α and INF-β.

    Viral ssRNA is sensed in the cell by RIG-I-like receptors (RLR), which causes ubiquitination of STING,

    which promotes the phosphorylation and activation of TRAF3.6 TRAF3 promotes activation of TBK1/IKKε

    which promotes NF-κB and activates IRF-3 dimerization, which that allows IRF-3 to enter the nucleus,

    where it is a transcription factor for interferon that is essential to mounting an adequate antiviral defense.7 8

    Both SARS-CoV and SARS-CoV-2 viruses code for a viral enzyme, Papain-like Protease (PLPro - EC

    3.4.22.B50). This protein inhibits the ubiquitination of STING, and the phosphorylation of TRAF3. Thus,

    the virus can defeat much of the TLR3 – TRIF – TRAF3 pathway for expression of interferon, and thereby

    inhibit the activation of NK cells and T cells and their production of antibodies to the virus. PLPro also

    interferes with the TLR7 signalling pathway.9 Viral protein inactivation of IFNα and IFNβ depress innate

    immunity and limit T-cell activation towards the formation of antibodies by B cells.

    Another SARS-CoV viral protein, ORF3a also known as Viroporin 3a, activates the NOD-like receptor P3

    (NRLP3) inflammasome. Viporin acts as an ion channel allowing K+ efflux and release of ROS from the

    mitochondria.10 The inflammasome activates Caspase 1, which cleaves pro-IL-1β into the active

    proinflammatory cytokine IL-1β.11 This action may be further upregulated by HMGB1 TLR2 activation.12

    ORF3a activates ACS and thus pyroptosis – thus can induce programmed cell death. Why would it be to

    virus’s evolutionary advantage to kill its host cell on which it is dependent for reproduction?

    It appears that SARS-CoV-2 can also infect lymphocytes. Although lymphocytes bear very low levels of

    ACE2, the virus appear to be able to enter these cells via CD147 (aka basigin).13 CD147 is essential for T-cell

    and NK cell development and differentiation and mediates the specific type of immune target these cells

    recognize.14 Thus, the coronavirus may cause lymphopenia by inducing pyroptosis in lymphocytes or

    common lymphoid precursor cells. Viral ORF3a may thus help eliminate Natural Killer cells and T cells

    essential for anti-viral defense.

    Red blood cells bear very high levels of CD147, however, these cells lack the cellular machinery to reproduce

    viruses.

  • (Adapted from KEGG Influenza pathway, modified to reflect COVID and viral proteins PLPro and ORF3a)

    In a study of mice infected with SARS virus, mice with MyD88 knockout had a mortality rate over 90%

    within 6 days, while all the wild-type test mice survived the infection.15 Clearly the MyD88 pathway is

    critical to survival of this infection.

  • In a subsequent study, mice with TLR4, TLR3, or TRAM knockout mice, were infected with the SARS virus.

    They were found to lose more weight as a result of the infection than wild-type mice, but there were no

    deaths. When TRIF (toll-like receptor adapter molecule 1) knock-out mice were infected with the SARS virus

    they had reduced lung function, more lung injury, higher viral loads and higher mortality than wild-type

    mice. Thus the TRIF pathway is an important for survival from SARS COVID infections.16

    If both TRIF and MyD88 pathways are impaired, there is little way to fight these infections. SARS-CoV viral

    papain-like protease (PLPro) is a protein that impairs the adaptive immune system, by inhibiting the

    formation of interferon alpha and beta. If the virus can defeat the TRIF pathway at the level of TRAF3 via its

    enzyme PLPro,17 that leaves the NFκB pathways. As a result of an incompetent response to the virus and

    inability to adequately mount T cell activation and antibody response, there is then a cycling upregulation of

    IL-1, IL-6, TNFα, and other cytokines. The recruitment of monocytes and macrophages causes tissue injury

    and activation of the NF-κB activates JAK/STAT1 and activation of the inflammasome, which cause the

    release of HMGB1. HMGB1 activates and reinforces TLR4, creating a positive feedback loop of cytokine

    release, inflammations, macrophage recruitment, and injury but without activation of NK and T-cells.

    It can thus be understood that inhibiting the immune response early in the TLR3 and TLR4 pathways in

    viral target cells, wherein activation of MyD88 or TRIF are inhibited, it impedes the ability to signal for the

    development of an appropriate immune response. Production of interferon α and β, the activation of NK

    cells, and development of T1 helper cells and of antiviral antibodies are essential to mount a defense and

    recovery from SARS, influenza and COVID.

    TLR4 also mediates the MAPK signalling pathway and AP-1 which promote the chemotaxis and survival of

    white blood cells. AP-1 signally may worsen tissue damage in the lungs and endothelial cells. While some

    aspects of the pathway are essential to forming immunity, INF-β production from non-infected cells

    stimulated by DAMPs can stimulate the JAK/STAT1 signalling pathway, which further promotes HMGB1

    migration from the nucleus to the cytoplasm. Cell death also releases HMGB1, which like OxPL, activates

    TLR4. Thus HMGB1 can create a positive feedback that ramps up tissue injury. Thus, incomplete TLR4

    response in infected cells mutes the immune response and allows viral proliferation; excessive immune

    response promotes severe life-threatening damage by monocytes and macrophages in response to PAMPS

    such as HMGB1 later in the disease process.18 HMGB1 is a potent chemokine and activator of immune

    mediated cell destruction that plays an important role in the lung damage occurring in SARS, influenza and

    presumably COVID-19.

    Medication:

    The beta blocker labetalol has been identified as potentially having SARS-CoV viral papain-like protease

    (PLPro) enzyme inhibitory effects. 19 Thus, it may help prevent SARS-CoV-2 immune evasion. Additionally,

    labetalol lowers blood pressure and should protect the heart from arrhythmias caused by long QTc,20 (both

    common issues in patients with severe COVID-19) and thus may reduce risk of sudden cardiac death

    associated with this disease.

    I suggest a trial of labetalol in appropriate COVID-19 patients. I suggest a starting treatment dose at about

    150 mg/ day in divided doses for a 70 kg adult.

  • Cytokines in COVID-19

    In a Chinese study comparing severe and critical ICU COVID-19 patients to those with less serious illness,

    those with severed disease had higher blood levels of interleukin-2 (IL-2), IL-7, granulocyte-colony

    stimulating factor (GSCF), interferon-γ inducible protein 10 (IP10), monocyte chemoattractant protein 1

    (MCP1) (aka; CCL2)), macrophage inflammatory protein 1-α (MIP1α), and tumor necrosis factor-α

    (TNFα).21

    In a separate study of bronchoalveolar lavage fluid and monocytes of COVID-19 patients, CCL2 (MCP-1),

    CXCL10 (IP-10) CCL3 (MIP-1A) and CCL4 (MIP1B) were highly expressed.22

    In SARS-COV, the spike protein triggers ACE2 signalling, which activates the Ras →ERK→AP-1 pathway.

    TNFα transcription is induced by AP-1 as well as by other inflammatory transcription factors.

    TNFα strongly induces numerous inflammatory molecules. In breast cancer cells, exposure to TNFα was

    found to increase the transcription of CCL2 by over 60 times, CXCL10 by 29 times, and complement C3 by

    six times.23 TNFα strongly induces the expression of fibrosis-associated CCL2. CCL2 is a chemokine for

    monocytes and macrophages. Additionally, CCL2 limits the survival of antibody forming cells in the lymph

    nodes.24 CCL2 was associated with severe lung injury and fibrosis in SARS.25

    Treatment:

    Apigenin, a phenolic compound present in certain edible plants, inhibits the canonical and non-canonical

    NLRP3 inflammasome pathways by decreasing caspase-1 and caspase-11 enzyme expression and activity.

    Apigenin strongly down-regulates CCL2.26 In tumor cells, a non-toxic dose of apigenin was found to reduce

    TNFα induced transcription of IKBKε by 108%, MLKL (a mediator of the necrosome/inflammasome

    pathway) by 106%, CCL2 by 97%, complement C3 by 91%, IL-6 by 88%, CXCL10 by 83%, TLR2 by 79%,

    PAI-1 by 77%, the IL7 receptor by 76%, and Complement Factor B (CFB) by 68%.27 Apigenin upregulates

    miR-155.28 miR-155 inhibits TNFα and the NF-κB signaling pathway in cytokine storm in animal models

    and human cell macrophages.29 The down regulation of C3 and CFB by apigenin are of significant interest in

    this disease, as complement activation is associated with worse outcome in mice with SARS, as discussed

    below.

    Cathespin S (CAT S) expression was found to be increased six times by TNFα in cancer cells in the study

    above, and was down regulated by apigenin by 66%.27 CAT S is a lysosomal cysteine protease. This enzyme

    has an important role in antigen presentation; it cleaves proteins into little chucks so that they can be

    presented on MHC II (major histocompatibility complex II) molecules for display on the cell surface. This

    allows immune cells to tell if the cell is adorned with “self” proteins or foreign proteins – in which case the

    T-cells will attack the cell and promote the development of antibodies to the foreign antigen. If there is

    insufficient CAT S, then large pieces of the proteins are displayed on the MHC II; this decreases immune

    recognition of foreign proteins and immune response to them. If there is over-abundant CAT S, it can cause

    the proteins to be cut into small fragments and be presented by the MHC II; this can trigger autoimmunity.

    Thus, while up-regulation of TNFα assists in mounting an immune response, excessive levels can promote

    autoimmune injury.30 Perhaps during cytokine storm, non-infected cells, over stimulated by TNFα and over-

    producing CAT S, may become targets of attack by the immune system. Furthermore, CAT S from alveolar

    macrophages, a cysteine protease, can also act as elastase. Thus it may promote alveolar injury similar to

  • that caused by neutrophil elastase, which causes emphysema. This may be especially damaging during

    mechanical ventilation.31

    I recommend the use of apigenin for the prevention and treatment of COVID-19, cytokine storm, sepsis, and

    viral pneumonia, and herein, specifically for the prevention and treatment of severe COVID-19. The dose

    estimate for apigenin for a severely ill COVID-19 patient is about 250 to 400 mg per day, while 100 mg per

    day in divided doses maybe helpful for persons at high risk of severe COVID-19 associated with chronic

    endothelial disease (HTN, type 2 diabetes, CAD, etc.). Preferably apigenin should be used in combination

    with other agents, such as pomegranate juice, as discussed below.

    Coagulation/Complement Cascade

    In another mouse study of SARS-CoV, mice with complement C3 knock-out mice (C3-/-) were infected with

    the SARS virus. Wild-type (C3+/+) mice when infected with the virus generally have viral infection in the

    lungs, lose weight and have an increase in inflammatory cytokines, but do not die. With SARS infection C3-/-

    mice has less weight loss, reduced lung pathology and lower cytokine levels in both the lung and blood. The

    viral load in the lungs of the C3-/- mice was no different than in the C3+/+ mice. This suggests that the

    complement cascade while exacerbating the pathology of SARS provides little if any benefit in clearing the

    disease. Thus therapy that inhibits the complement cascade may be helpful in treating the disease.32

    There are multiple pathways for activation of the complement system; some of the pathways are closely

    aligned with coagulation. Microbes can activate C3, but it is also activated by plasmin and thrombin. When

    C3 becomes activated, it activates C5. Thrombin may activate C5 directly.

    Complement factor B mediates the alternative pathway activation of both C3 and C5. The alternative

    pathway is activated by infection. Apigenin down-regulates TNFα induced upregulation of C3 and CFB.

    In severe SARS-CoV infections there is diffuse alveolar damage, vascular leakage into the alveolar spaces,

    premature breakdown of fibrin clots and possible micro-hemorrhage in the lungs. Similar pathology is seen

    with severe strains of influenza, including the 1918 and 2009 H1N1 influenza viruses. In the 1918 H1N1

    pandemic, many young patients died a hemorrhagic death. With the outbreak of SARS in 2002, we were

    fortunate that the outbreak was small, and there were a limited number of cases. With COVID-19, were have

    found, that while presenting as a pulmonary disease with hypoxia, it likely primarily affects the endothelial

    cells of the vasculature, and microvasculature, causing vasoconstriction and micro emboli to the lungs.

    COVID-19 patients with elevated levels of D-dimer, a fibrin degradation product, have a higher mortality.33

    D-dimer is an indicator that there has been clot formation and breakdown. Heparin and warfarin inhibit

    coagulation Factor Xa, an enzyme that promotes cleavage of prothrombin to thrombin, which activates C3.

    Heparin therapy in COVID-19 patients appears to lower mortality.

    A series of autopsy of four COVID-19 cases was performed at Tulane University Medical Center. All the

    decedents had had elevated ferritin, fibrinogen, PT (prothrombin time), and very elevated D-dimer,

    indicating disseminated intravascular coagulation. Nevertheless, on autopsy, thrombotic microangiopathy

  • was restricted to the lungs. The lungs were heavy (wet) lungs and the decedents had dilated right heart

    ventricle, suggestive of right heart failure following pulmonary hypertension and pulmonary edema. There

    was an abundance of megakaryocytes in the lungs, as had been found with SARS infections. These cells

    produce platelets, and likely play a role in micro-thrombosis and blockage of the pulmonary capillary bed. 34

    In early disease, such pulmonary capillary blockage would cause the shunting of blood through un-aerated

    parts of the lung, causing hypoxia. This would be exacerbated by vasoconstriction. As the disease

    progressed, the elevated capillary pressure would cause leakage of fluid into the intestinal fluid and into the

    alveoli causing pulmonary edema and worsening hypoxia. Eventually, the right ventricle of the heart may

    become unable to pump sufficient blood through the lungs and suffer exhaustion and fail.

    There is renal injury in severe COVID-19. More than half of hospitalized COVID-19 patients with severe not

    prior history of kidney disease were found to have proteinuria, hematuria and leukocyturia.35 Many COVID-

    19 survivors require dialysis.

  • The coagulation/complement cascades is likely important in the causation of the “dry” lung manifestation of COVID-19. Virally induced endothelial injury in blood vessels causes oxidative injury, los of nitric oxide production and thus vasoconstriction, platelet activation, activation of prothrombin to thrombin as a result of activation of Factor X to Factor Xa. This causes the formation of microemboli that are filtered in the capillaries of the lung. This would explain the hypoxia with normal lung compliance seen in the dry manifestation of the disease. Heparin inhibits the enzymatic activity of Factor Xa.

    As illustrated above, the coagulation cascade promotes activation of the complement system via both

    thrombin (pro-fibrin production) and plasmin (pro-fibrinolysis). The complement system is part of the

    innate immune system. It promotes chemotaxis, drawing more white blood cells to the area and promotes

    degranulation and phagocytosis. C5 activation (by C3) promotes assembly of the Membrane Attack

    Complex, which targets and kills virally infected cells; in COVID-19, the alveolar cells are the target.

    Activation of the complement cascade occurs as early as the first day following inoculation of animals with

    the SARS-CoV virus. Complement activation is known to increased vascular permeability, a feature of severe

    SARS-CoV infection that is associated with poor outcome. Baseline complement activity increases with age,

    and is consistent with the increase mortality with age in SARS. Furthermore, complement activation is

    predictive of the development of ARDS. Complement activation increases inflammation and promotes lysis

    of cells, causing the release of damage associated molecular patterns, (DAMPs) such as HMGB1. DAMPS

    can then further activate the inflammatory cascade and the complement system.36

    IL-1β and TNFα → uPA → Plasmin → C3 → C3a, C3b and C5 activation

    DAMPs → C1, C4 → C3 → C3a, C3b and C5 activation

    Viral endothelial injury → X → Xa → Thombin → C3

    Tissue plasminogen activator (tPA) (EC 3.4.21.68) also activates plasminogen as does uPA. Kallikrein also

    promotes plasminogen.37 Later in the disease course of severe SARS, there is fibrin degradation and

    bleeding into the alveoli. This may be followed by lung fibrosis in survivors.

    Medications:

    Anti-coagulation therapy likely needs to be part of the treatment of regimen of hospitalized patients with

    COVID-19. Acetylsalicylic acid, P2Y12 inhibitors, and glycoprotein IIb/IIIa antagonists, may reduce lung

    injury and mortality in COVID without increased bleeding risk.38 Nevertheless, many studies have failed to

    find a survival benefit using anticoagulant therapy in sepsis.39

    As shown in the figure above, heparin (LMWH) is a logical choice in the treatment of severe COVID-19 for

    the prevention of coagulopathy and complement activation.

    Statin drugs also impact the PAI-1 and tPA. Statins induce tPA and inhibits plasminogen activator inhibitor-

    1. Thus, statins have an anti-fibrosis effect on wound healing by promoting the degradation of fibrin

  • products.40 41 Atorvastatin has been found to increase the expression of ACE2 in the heart of animals.42 43

    This may or may not be a class effect. Statins also downregulate ICAM-1, a protein whose expression is

    required for Killer T-cells to adhere and eliminate viral-infected cells.44 I suggest that simvastatin and other

    statin medications be avoided during the course of COVID. If they are used, I suggest they be used at very

    low dose, i.e.; 5 mg of simvastatin per day.

    Urokinase plasminogen activator (uPA) has been found to play a central activating role in causing fibrosis

    and lung injury in SARS.45 uPA activates the conversion of plasminogen to plasmin which also activates C3

    via activation of plasminogen to plasmin. uPA also activates uPAR (plasminogen activator urokinase

    receptor) which induces cell adhesion, migration, and proliferation.46 The inflammatory cytokines IL-1β 47

    and TNF-α 48 induce the activation/expression of uPA.

    A literature search for readily available medications that inhibit uPA revealed amiloride as a potential

    candidate. 49 This is an anti-hypertensive drug, and this seems to be a unique feature of this medication. This

    medicine may also inhibit tissue kallikrein,50 an enzyme that upregulates plasminogen.51 This is not the

    drug’s mechanism of action as an anti-hypertensive medication, but rather a side effect that may be useful in

    treating ARDS in SARS and severe influenza. It may reduce injury and help prevent late effects of the

    disease.

    In COVID-19, the development of hypertension is part of the disease pathology in severe disease. Thus

    treatment is often needed for hypertension and congestive heart failure in severely and critically ill COVID

    patients. Amiloride is a potassium-sparing diuretic. Hypokalemia has been found in 93 percent of patients

    with severe or critical COVID-19, as a result of urinary loss of potassium resulting from degradation of

    ACE2.52 Amiloride may decrease potassium loss; however, potassium levels will still need to be monitored. I

    suggest a trial using low-dose amiloride (2.5 mg/day) for its anti-uPA effect. Higher doses may have adverse

    effects, especially if the patient is not anticoagulated.

    HMGB1

    HMGB1 is a moderator of inflammation and survival of phagocytic white blood cells, and causes prolonged,

    vigorous tissue destruction in influenza and SARS, and likely COVID-19. HMGB1 is a key pathway in sepsis

    and septic shock and mediates organ damage. HMGB1 is a nuclear protein that leaks into the cytoplasm in

    inflammation and the out of the cell in cell injury of cell death. In cancer, HMGB1 promotes cancer cell

    proliferation, migration and tissue invasion. It is a DAMP (damage-associated molecular pattern) that

    activates TLR4 (and TLR2) and induces inflammatory cytokines in cytokine storm. DAMPs create an

    inflammatory cascade (HMGB1/TLR4/ MyD88/NF-κB) that can result in tissue destruction.

    Hypoxia/reperfusion, oxygen glucose deprivation, oxidative stress, and other stressors can cause the

    acetylation of the nuclear HMGB1 promoting its translocation into the cytosol and then into the

    extracellular space. Pathogen-associated molecular pattern (PAMP) molecules, such as LPS and Ox-PL,

    which is released in viral infections such as influenza A and SARS, can activate the TLR4 pathway. The

    TLR4 pathway can activate the JAK/STAT1 pathway that also promotes the acetylation and translocation of

    HMGB1 from the cell’s nucleus to the cytoplasm.

  • Pyroptosis, a form of induced programmed cell death that is part of the innate immune response to protect

    against intracellular microbial infection. In pyroptosis there is inhibition of internal pathogen reproduction

    and promotion of phagocytosis. Pyroptosis involves TLRs, Casp1 activation the inflammasome and NOD-

    like receptors. Extracellular ATP, oxidized mitochondrial DNA, mitochondrial ROS, and leakage of

    cardiolipin from the mitochondria together promote the activation of the NRLP3 inflammasome, which

    activates pro-caspase-1 to Casp-1. Casp-1 activates pro-IL1β and pro-IL-18 to IL1β and IL-18. TLR4 can also

    induce IL-1β processing via Casp-8. Activation of the TNF receptor promotes pro-IL-1β and NLRP3

    inflammasome transcription via nuclear factor-κB (NF-κB) activation.

    MicroRNAs (miR) are non-coding small hairpin-shaped sections of RNA that bind to specific mRNAs, and

    prevent the mRNA from moving through the ribosome. They thus inhibit the production of the protein for

    that the mRNA translates. This is an important way the cell stops the production of various proteins when

    there is a sufficient supply or when there is no further need for that protein. Many medications act by

    inhibiting enzymes or inhibiting or activation cell receptors. Other proteins are considered non-drugable.

    Promoting or inhibiting the production of miR may allow pharmacologic control of some traditionally “non-

    drugable” proteins and pathways. Thus, I explored miRNA induction as a potential treatment of virally

    induced cytokine storm.

    Several miR have been identified that, some experimentally in cell culture and others by chemical modeling,

    that down-regulate HMGB1 protein translation. Additionally, several compounds have been identified that

    either up or down-regulate the production of miR that reduce HMGB1 activity. HMGB1 promotes an

    increase in the expression of miR-21 and miR-129-2, and some of its effects are mediated by these

    microRNA. It may be possible to intervene by inhibiting these. Certain miR have also been identified to be

    in higher concentration in patients with cytokine storm or sepsis, and these may provide clues to treatment,

    however, with this type of data, it is essential to understand whether the rise is part of the cause of injury,

    part of the response to injury.

    Human pulmonary/cardiac microvascular endothelial cells (HMVECs) are available that can be grown in

    cell culture. Cultured HMVECs grown in the presence of thrombin have increased permeability. Over-

    expression of miR-126 decreased endothelial space, thus preventing he increase in permeability.53 In mice,

    over-expression of miR-126 decreased serum levels of IL-6 and TNF-α in a model of sepsis, and lowered the

    mortality rate. In humans, miR-126 has been found to be down-regulated in patients with hospitalized with

    sepsis, and the degree of down-regulation correlated to the severity of sepsis early in the disease

    progression.54 In a separate study, miR-126 was found to protect HCMECs against hypoxia/reoxygenation

    injury and to decrease inflammatory Reactive oxygen species ROS, and reduce the expression of IL6, IL10

    and TNFα, while increasing the vasodilator NO, and the antioxidant SOD.55

    The table below summarizes the effect of several microRNA on HMGB1 and cytokine storm relevant to

    COVID-19, and list some agents that impact the expression of these miR. Those estimated to be most

    important in COVID-19 and are highlighted in bold. Note that that the medications metformin and the PPI,

    esomeprazole as well as carnosic acid inhibit the expression of miR that reduce HMGB1 translation and thus may worsen COVID-19 disease. All PPIs may have this effect. Olive oil and vitamin D deficiency may

    increase HMGB1 activity, and thus perhaps should also be avoided.

    Pomegranate Juice: In a gene assay, pomegranate juice up-regulated genes involved in cell adhesion

    such as E-cadherin, intercellular adhesion molecule 1 (ICAM-1) and down-regulates genes involved in cell

  • migration such as hyaluranan-mediated motility receptor (HMMR) and type I collagen. It up-regulated

    microRNAs including miR-335, miR-205, miR-200, and miR-126, and down-regulates miR-21 and miR-

    373. Pomegranate juice reduces the level of pro-inflammatory cytokines/chemokines such as IL-6, IL-

    12p40, IL-1β and RANTES. 56 In an in vivo study, rats receiving pomegranate juice had significantly down-

    regulated proinflammatory enzymes nitric oxide synthase and cyclooxygenase-2 messenger RNA (mRNA)

    and protein expression. NF-κB and VCAM-1 mRNA and proteins expression were suppressed. There was

    also inhibition of phosphorylation of PI3K/AKT and mTOR expression and increased the expression of miR-

    126.57 Pomegranate juice also is expected to down-regulates HMGB1 via miR-200a and likely by miR-20558

    and also reduces HMGB1 downstream activity as a result of reduced expression of miR-21 and let-7c. 59

    I recommend the use of pomegranate juice (but not other pomegranate extracts) for the prevention and

    treatment of cytokine storm, sepsis, and viral pneumonia, and herein, specifically for the prevention and

    treatment of severe COVID19. I suggest trials beginning with 4 to 8 ounces of pomegranate juice or about 4

    to 10 grams of freeze dried pomegranate juice powder per day; higher end doses may be required for more

    severe disease and larger patients.

  • miRNA and HMGB1 60 61 62 63 64 65 66 67 68 69

    miRNA MicroRNA Effect Upregulates (Desirable)

    Downregulates (Undesirable)

    miR-126 Decreases serum IL-6 and TNF-α in sepsis, decreased intracellular permeability in cell culture. Protects from hypoxia/re-oxygenation/ reperfusion injury

    Pomegranate juice70 Mango polyphenols71 72

    miR-129-5p Suppresses HMGB1, RAGE Metformin Downregulated by 40%73

    miR-193a-3p74 Targets HMGB1, TGF-β, and HYOU1 Resveratrol

    Metformin lowers by 55%75

    mir-200a Lowers HMGB1 expression Pomegranate Juice

    miR-34a Suppresses HMGB1 (Downregulated in septic shock)

    Honokiol, resveratrol and n3-PUFA Pomegranate rind76 EGCG

    Carnosic acid77 (Present in rosemary and sage) Quercetin Curcumin

    miR-376a Lowers HMGB1 expression Esomeprazole, (proton pump inhibitors)78

    miR-15579 Inhibits the NF-κB signaling pathway and TNFα.

    Apigenin80 Resveratrol81

    Quercitin82 Pomegranate

    extract

    miR-320a, miR-325 and miR-505

    Lower HMGB1 expression

    miRNA MicroRNA Effect Upregulates (Undesirable)

    Downregulates (Desirable)

    miR-21 83 HMGB1 promotes miR-21 and NF-κB pathway activity

    Oleic acid (olive oil) DIM, Choline, and Folic acid up-regulate. Curcumin Vit. D3 deficiency n-3 fatty acid deficiency

    Resveratrol Avoid vitamin D3 and n-3 fatty acid deficiencies

    miR-129-2 HMGB1 increases

    let-7c HMGB1 increases EGCG Quercetin curcumin

    Pomegranate Juice, Resveratrol

    Note: Not intended to be a comprehensive listing of miRNA related activity, but rather list those

    encountered that are pertinent to HMGB1 and cytokine storm in viral pneumonia and COVID-19.

  • Summary

    Medications that may be helpful for COVID-19 and similar cytokine storm, DIC, sepsis conditions, dose

    estimate per day for a 70 kg adult:

    1. Low molecular weight heparin (LMWH) All treatment patients should be heparinized and monitored

    with aPTT as per usual hospital protocol. This may already be part of the hospital’s usual care for

    COVID patients. I recommend that all patients (test and control patients) without contraindication

    be placed on LMWH.

    2. Labetalol 150 mg (75 mg/BID)

    3. Amiloride 2.5 mg QD

    4. Vitamin D3 500 IU QD

    Natural Agents that may be helpful in the prevention or treatment of severe COVID with daily dose. In

    addition to those agents already discussed, I also recommend ginger juice as an antiviral agent for use in

    COVID infection.

    Divided into 4 or five doses per day:

    1. Pomegranate Juice: 3 ml/kg day of juice or 100 mg of powder per Kg Juice Source Powder Source

    2. Apigenin 250 mg Source

    Notes:

    Freeze dried pomegranate juice may be used as a substituted for pomegranate juice, using about 1.5 grams

    per ounce of juice.

    A small dose of vitamin D3 is recommended daily as 25 hydroxyvitamin D3 is required for new WBC

    function as new cells are created daily, but the body stores vitamin D as 1-25 hydroxyvitamin D3. Studies of

    vitamin D3 use in influenza support the use of low doses, but not higher doses.

    I am unaware of any other contraindication to using these medications together or together with the natural

    agents listed above.

    I also recommend avoidance of statin medications, proton pump inhibitors, and metformin during the

    course of this disease, as well as avoidance of olive oil, sage, rosemary and curcumin/turmeric as a result of

    their miRNA activity on HMGB1. Since oleic acid may remain in the body for an extended time, it may be

    prudent to avoid it if there is high risk of COVID infection morbidity.

    These agents are not intended to displace of other anti-SARS-CoV-2 medications. Antiviral agents such as

    Camostat mesilate84 or remdesivir may be helpful, especially early in the disease. The focus here in is to

    improve the immune response to the disease.

    https://www.amazon.com/POM-Wonderful-Pomegranate-Juice-Count/dp/B004O4BB4Whttps://www.amazon.com/Navitas-Organics-Pomegranate-Powder-oz/dp/B001TNW23Uhttps://www.amazon.com/Swanson-Apigenin-Prostate-Supplements-Capsules/dp/B001TEIJIQ/

  • Additional notes:

    TMPRSS2, the host cell protein on cells that that mediates uptake of the SARS-CoV-2 virus after binding to

    ACD2, is inhibited by plasminogen activator inhibitor-1 (PAI-1).85 PAI-1 in inhibited by statin drugs.

    TMPRSS2 is even more strongly inhibited by antithrombin, which is activated by heparin86 in physiologic

    levels of calcium. PAI-1 expression greatly augmented by TNF-α, and this greatly down-regulated by

    apigenin.

    Apigenin is a Src-tyrosine kinase inhibitor that inhibits activation of SHP1 and SHP2, thus inhibiting the

    IDO mediated downregulation of Th1 and NK cell immune activity. 87

    Apigenin reduces NLRP3 inflammasome activation.88

    Please let me know if tested in patients. Thanks dr.charles.lewisgmail.com

    1 Severe acute respiratory syndrome coronavirus ORF3a protein activates the NLRP3 inflammasome by promoting TRAF3-dependent ubiquitination of ASC. Siu KL, Yuen KS, Castaño-Rodriguez C, Ye ZW, Yeung ML, Fung SY, Yuan S, Chan CP, Yuen KY, Enjuanes L, Jin DY. FASEB J. 2019 Aug;33(8):8865-8877. doi: 10.1096/fj.201802418R. PMID:31034780

    2 Immune responses in COVID-19 and potential vaccines: Lessons learned from SARS and MERS epidemic. Prompetchara E, Ketloy C, Palaga T. Asian Pac J Allergy Immunol. 2020 Mar;38(1):1-9. doi: 10.12932/AP-200220-0772. PMID:32105090

    3 COVID-19 pneumonia: different respiratory treatment for different phenotypes?L.Gattinoni et al. https://www.esicm.org/wp-content/uploads/2020/04/684_author-proof.pdf

    4 Identification of oxidative stress and Toll-like receptor 4 signaling as a key pathway of acute lung injury. Imai Y, Kuba K, Neely GG, Yaghubian-Malhami R, Perkmann T, van Loo G, Ermolaeva M, Veldhuizen R, Leung YH, Wang H, Liu H, Sun Y, Pasparakis M, Kopf M, Mech C, Bavari S, Peiris JS, Slutsky AS, Akira S, Hultqvist M, Holmdahl R, Nicholls J, Jiang C, Binder CJ, Penninger JM. Cell. 2008 Apr 18;133(2):235-49. doi: 10.1016/j.cell.2008.02.043. PMID:18423196

    5 The TLR4-TRIF pathway protects against H5N1 influenza virus infection. Shinya K, Ito M, Makino A, Tanaka M, Miyake K, Eisfeld AJ, Kawaoka Y. J Virol. 2012 Jan;86(1):19-24. doi: 10.1128/JVI.06168-11. PMID:22031950

    6 cGAS and STING: At the intersection of DNA and RNA virus-sensing networks. Ni G, Ma Z, Damania B. PLoS Pathog. 2018 Aug 16;14(8):e1007148. doi: 10.1371/journal.ppat.1007148. PMID:30114241

    7 Coronavirus papain-like proteases negatively regulate antiviral innate immune response through disruption of STING-mediated signaling. Sun L, Xing Y, Chen X, Zheng Y, Yang Y, Nichols DB, Clementz MA, Banach BS, Li K, Baker SC, Chen Z. PLoS One. 2012;7(2):e30802. doi: 10.1371/journal.pone.0030802. PMID:22312431

    https://www.ncbi.nlm.nih.gov/pubmed/31034780https://www.ncbi.nlm.nih.gov/pubmed/31034780https://www.ncbi.nlm.nih.gov/pubmed/32105090https://www.esicm.org/wp-content/uploads/2020/04/684_author-proof.pdfhttps://www.ncbi.nlm.nih.gov/pubmed/18423196https://www.ncbi.nlm.nih.gov/pubmed/22031950https://www.ncbi.nlm.nih.gov/pubmed/30114241https://www.ncbi.nlm.nih.gov/pubmed/22312431https://www.ncbi.nlm.nih.gov/pubmed/22312431

  • 8 SARS coronavirus papain-like protease inhibits the type I interferon signaling pathway through interaction with the STING-TRAF3-TBK1 complex. Chen X, Yang X, Zheng Y, Yang Y, Xing Y, Chen Z. Protein Cell. 2014 May;5(5):369-81. doi: 10.1007/s13238-014-0026-3. PMID:24622840

    9 SARS Coronavirus Papain-Like Protease Inhibits the TLR7 Signaling Pathway through Removing Lys63-Linked Polyubiquitination of TRAF3 and TRAF6. Li SW, Wang CY, Jou YJ, Huang SH, Hsiao LH, Wan L, Lin YJ, Kung SH, Lin CW. Int J Mol Sci. 2016 May 5;17(5). pii: E678. doi: 10.3390/ijms17050678. PMID:27164085

    10 Severe Acute Respiratory Syndrome Coronavirus Viroporin 3a Activates the NLRP3 Inflammasome. Chen IY, Moriyama M, Chang MF, Ichinohe T. Front Microbiol. 2019 Jan 29;10:50. doi: 10.3389/fmicb.2019.00050. PMID:30761102

    11 Mitochondrial protein mitofusin 2 is required for NLRP3 inflammasome activation after RNA virus infection. Ichinohe T, Yamazaki T, Koshiba T, Yanagi Y. Proc Natl Acad Sci U S A. 2013 Oct 29;110(44):17963-8. doi: 10.1073/pnas.1312571110. PMID:24127597

    12 TLR2/MyD88/NF-κB pathway, reactive oxygen species, potassium efflux activates NLRP3/ASC inflammasome during respiratory syncytial virus infection. Segovia J, Sabbah A, Mgbemena V, Tsai SY, Chang TH, Berton MT, Morris IR, Allen IC, Ting JP, Bose S. PLoS One. 2012;7(1):e29695. doi: 10.1371/journal.pone.0029695. PMID:22295065

    13 SARS-CoV-2 invades host cells via a novel route: CD147-spike protein. Wang. K, et al. 3/14/2020 https://www.biorxiv.org/content/10.1101/2020.03.14.988345v1.

    14 Targeting CD147 for T to NK Lineage Reprogramming and Tumor Therapy. Geng JJ, Tang J, Yang XM, Chen R, Zhang Y, Zhang K, Miao JL, Chen ZN, Zhu P. EBioMedicine. 2017 Jun;20:98-108. doi: 10.1016/j.ebiom.2017.05.022. PMID:28571672

    15 MyD88 is required for protection from lethal infection with a mouse-adapted SARS-CoV. Sheahan T, Morrison TE, Funkhouser W, Uematsu S, Akira S, Baric RS, Heise MT. PLoS Pathog. 2008 Dec;4(12):e1000240. doi: 10.1371/journal.ppat.1000240. PMID:19079579

    16 Toll-Like Receptor 3 Signaling via TRIF Contributes to a Protective Innate Immune Response to Severe Acute Respiratory Syndrome Coronavirus Infection. Totura AL, Whitmore A, Agnihothram S, Schäfer A, Katze MG, Heise MT, Baric RS. mBio. 2015 May 26;6(3):e00638-15. doi: 10.1128/mBio.00638-15. PMID:26015500

    17 SARS coronavirus papain-like protease inhibits the type I interferon signaling pathway through interaction with the STING-TRAF3-TBK1 complex. Chen X, Yang X, Zheng Y, Yang Y, Xing Y, Chen Z. Protein Cell. 2014 May;5(5):369-81. doi: 10.1007/s13238-014-0026-3. PMID:24622840

    18 Toll-like receptor 4 in acute viral infection: Too much of a good thing. Olejnik J, Hume AJ, Mühlberger E. PLoS Pathog. 2018 Dec 20;14(12):e1007390. doi: 10.1371/journal.ppat.1007390. eCollection 2018 Dec. PMID:30571771

    19 Potential inhibitors against papain-like protease of novel coronavirus (SARS-CoV-2) from FDA approved drugs. https://chemrxiv.org/articles/Potential_Inhibitors_Against_Papain-like_Protease_of_Novel_Coronavirus_COVID-19_from_FDA_Approved_Drugs/11860011

    https://www.ncbi.nlm.nih.gov/pubmed/24622840https://www.ncbi.nlm.nih.gov/pubmed/24622840https://www.ncbi.nlm.nih.gov/pubmed/27164085https://www.ncbi.nlm.nih.gov/pubmed/27164085https://www.ncbi.nlm.nih.gov/pubmed/30761102https://www.ncbi.nlm.nih.gov/pubmed/24127597https://www.ncbi.nlm.nih.gov/pubmed/24127597https://www.ncbi.nlm.nih.gov/pubmed/22295065https://www.ncbi.nlm.nih.gov/pubmed/22295065https://www.biorxiv.org/content/10.1101/2020.03.14.988345v1https://www.ncbi.nlm.nih.gov/pubmed/28571672https://www.ncbi.nlm.nih.gov/pubmed/19079579https://www.ncbi.nlm.nih.gov/pubmed/26015500https://www.ncbi.nlm.nih.gov/pubmed/26015500https://www.ncbi.nlm.nih.gov/pubmed/24622840https://www.ncbi.nlm.nih.gov/pubmed/24622840https://www.ncbi.nlm.nih.gov/pubmed/30571771https://chemrxiv.org/articles/Potential_Inhibitors_Against_Papain-like_Protease_of_Novel_Coronavirus_COVID-19_from_FDA_Approved_Drugs/11860011https://chemrxiv.org/articles/Potential_Inhibitors_Against_Papain-like_Protease_of_Novel_Coronavirus_COVID-19_from_FDA_Approved_Drugs/11860011

  • 20 Role of alpha1-blockade in congenital long QT syndrome: investigation by exercise stress test. Furushima H, Chinushi M, Washizuka T, Aizawa Y. Jpn Circ J. 2001 Jul;65(7):654-8. PMID:11446501

    21 Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, Zhang L, Fan G, Xu J, Gu X, Cheng Z, Yu T, Xia J, Wei Y, Wu W, Xie X, Yin W, Li H, Liu M, Xiao Y, Gao H, Guo L, Xie J, Wang G, Jiang R, Gao Z, Jin Q, Wang J, Cao B. Lancet. 2020 Feb 15;395(10223):497-506. doi: 10.1016/S0140-6736(20)30183-5. PMID:31986264

    22 Transcriptomic characteristics of bronchoalveolar lavage fluid and peripheral blood mononuclear cells in COVID-19 patients. Xiong Y, Liu Y, Cao L, Wang D, Guo M, Jiang A, Guo D, Hu W, Yang J, Tang Z, Wu H, Lin Y, Zhang M, Zhang Q, Shi M, Liu Y, Zhou Y, Lan K, Chen Y. Emerg Microbes Infect. 2020 Dec;9(1):761-770. doi: 10.1080/22221751.2020.1747363. PMID:32228226

    23 Effect of apigenin on whole transcriptome profile of TNFα-activated MDA-MB-468 triple negative breast cancer cells. Bauer D, Mazzio E, Hilliard A, Oriaku ET, Soliman KFA. Oncol Lett. 2020 Mar;19(3):2123-2132. doi: 10.3892/ol.2020.11327. PMID:32194710

    24 https://immunology.sciencemag.org/content/5/45/eaaw0693

    25 Upregulation of the chemokine (C-C motif) ligand 2 via a severe acute respiratory syndrome coronavirus spike-ACE2 signaling pathway. Chen IY, Chang SC, Wu HY, Yu TC, Wei WC, Lin S, Chien CL, Chang MF. J Virol. 2010 Aug;84(15):7703-12. doi: 10.1128/JVI.02560-09. PMID:20484496

    26 Apigenin inhibits TNFα/IL-1α-induced CCL2 release through IKBK-epsilon signaling in MDA-MB-231 human breast cancer cells. Bauer D, Redmon N, Mazzio E, Soliman KF. PLoS One. 2017 Apr 25;12(4):e0175558. doi: 10.1371/journal.pone.0175558. PMID:28441391

    27 Whole Transcriptomic Analysis of Apigenin on TNFα Immuno-activated MDA-MB-231 Breast Cancer Cells. Bauer D, Mazzio E, Soliman KFA. Cancer Genomics Proteomics. 2019 Nov-Dec;16(6):421-431. doi: 10.21873/cgp.20146. PMID:31659097

    28 Dietary apigenin reduces LPS-induced expression of miR-155 restoring immune balance during inflammation. Arango D, Diosa-Toro M, Rojas-Hernandez LS, Cooperstone JL, Schwartz SJ, Mo X, Jiang J, Schmittgen TD, Doseff AI. Mol Nutr Food Res. 2015 Apr;59(4):763-72. doi: 10.1002/mnfr.201400705. PMID:25641956

    29 Cross-Regulation of Proinflammatory Cytokines by Interleukin-10 and miR-155 in Orientia tsutsugamushi-Infected Human Macrophages Prevents Cytokine Storm. Tsai MH, Chang CH, Tsai RK, Hong YR, Chuang TH, Fan KT, Peng CW, Wu CY, Hsu WL, Wang LS, Chen LK, Yu HS. J Invest Dermatol. 2016 Jul;136(7):1398-1407. doi: 10.1016/j.jid.2015.11.034. PMID:26921773

    30 https://en.wikipedia.org/wiki/Cathepsin_S

    31 Inflammatory Cellular Response to Mechanical Ventilation in Elastase-Induced Experimental Emphysema: Role of Preexisting Alveolar Macrophages Infiltration. Rouze A, Voiriot G, Guivarch E, Roux F, Tran Van Nhieu J, Isabey D, Brochard L, Maitre B, Mekontso-Dessap A, Boczkowski J. Biomed Res Int. 2018 Dec 19;2018:5721293. doi: 10.1155/2018/5721293. PMID:30662910

    https://www.ncbi.nlm.nih.gov/pubmed/11446501https://www.ncbi.nlm.nih.gov/pubmed/31986264https://www.ncbi.nlm.nih.gov/pubmed/32228226https://www.ncbi.nlm.nih.gov/pubmed/32228226https://www.ncbi.nlm.nih.gov/pubmed/32194710https://www.ncbi.nlm.nih.gov/pubmed/32194710https://immunology.sciencemag.org/content/5/45/eaaw0693https://www.ncbi.nlm.nih.gov/pubmed/20484496https://www.ncbi.nlm.nih.gov/pubmed/20484496https://www.ncbi.nlm.nih.gov/pubmed/28441391https://www.ncbi.nlm.nih.gov/pubmed/28441391https://www.ncbi.nlm.nih.gov/pubmed/31659097https://www.ncbi.nlm.nih.gov/pubmed/31659097https://www.ncbi.nlm.nih.gov/pubmed/25641956https://www.ncbi.nlm.nih.gov/pubmed/25641956https://www.ncbi.nlm.nih.gov/pubmed/26921773https://www.ncbi.nlm.nih.gov/pubmed/26921773https://en.wikipedia.org/wiki/Cathepsin_Shttps://www.ncbi.nlm.nih.gov/pubmed/30662910https://www.ncbi.nlm.nih.gov/pubmed/30662910

  • 32 Complement Activation Contributes to Severe Acute Respiratory Syndrome Coronavirus Pathogenesis. Gralinski LE, Sheahan TP, Morrison TE, Menachery VD, Jensen K, Leist SR, Whitmore A, Heise MT, Baric RS. mBio. 2018 Oct 9;9(5). pii: e01753-18. doi: 10.1128/mBio.01753-18. PMID:30301856

    33 Anticoagulation Guidance Emerging for Severe COVID-19 Phend, C. 5/8/2020 https://www.medpagetoday.com/infectiousdisease/covid19/85865 34 Pulmonary and Cardiac Pathology in Covid-19: The First Autopsy Series from New Orleans. Sharon E. Fox, Aibek Akmatbekov, Jack L. Harbert, Guang Li, J. Quincy Brown, Richard S. Vander Heide. doi: https://doi.org/10.1101/2020.04.06.20050575. Posted 5/10/2020 https://www.medrxiv.org/content/10.1101/2020.04.06.20050575v1

    35 Urinalysis, but not blood biochemistry, detects the early renal-impairment in patients with COVID-19. Zhou, H et al. https://www.medrxiv.org/content/10.1101/2020.04.03.20051722v1

    36 Complement, thrombotic microangiopathy and disseminated intravascular coagulation. Kurosawa S, Stearns-Kurosawa DJ. J Intensive Care. 2014 Dec 31;2(1):65. doi: 10.1186/s40560-014-0061-4. eCollection 2014. PMID:25705421

    37 https://www.kegg.jp/kegg-bin/show_pathway?map04610

    38 Coagulation abnormalities in sepsis. Tsao CM, Ho ST, Wu CC. Acta Anaesthesiol Taiwan. 2015 Mar;53(1):16-22. doi: 10.1016/j.aat.2014.11.002. PMID:25544351

    39 Efficacy and safety of anticoagulant therapy in three specific populations with sepsis: a meta-analysis of randomized controlled trials. Umemura Y, Yamakawa K, Ogura H, Yuhara H, Fujimi S. J Thromb Haemost. 2016 Mar;14(3):518-30. doi: 10.1111/jth.13230. PMID:26670422

    40 Simvastatin increases fibrinolytic activity in human peritoneal mesothelial cells independent of

    cholesterol lowering. Haslinger B. Kidney Int. 2002 PMID: 12371961

    41 Effect of statins on experimental postoperative adhesion: a systematic review and meta-analysis. Choi GJ, Park HK, Kim DS, Lee D, Kang H. Sci Rep. 2018 Oct 3;8(1):14754. doi: 10.1038/s41598-018-33145-z. PMID:30283040

    42 Tissue specific up regulation of ACE2 in rabbit model of atherosclerosis by atorvastatin: role of epigenetic histone modifications. Tikoo K, Patel G, Kumar S, Karpe PA, Sanghavi M, Malek V, Srinivasan K. Biochem Pharmacol. 2015 Feb 1;93(3):343-51. doi: 10.1016/j.bcp.2014.11.013. PMID:25482567

    43 [Effect of atorvastatin on ACE2 expression in pressure overload induced cardiac hypertrophy in rats]. Qin XT, Jia CW, Pan M, Shen AG, Jing HM. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2008 May;33(5):438-42. PMID:18544849

    44 Simvastatin Improves Cardiac Hypertrophy in Diabetic Rats by Attenuation of Oxidative Stress and Inflammation Induced by Calpain-1-Mediated Activation of Nuclear Factor-κB (NF-κB). Han Q, Liu Q, Zhang H, Lu M, Wang H, Tang F, Zhang Y. Med Sci Monit. 2019 Feb 15;25:1232-1241. doi: 10.12659/MSM.913244. PMID:30767945

    https://www.ncbi.nlm.nih.gov/pubmed/30301856https://www.medpagetoday.com/infectiousdisease/covid19/85865https://doi.org/10.1101/2020.04.06.20050575https://www.medrxiv.org/content/10.1101/2020.04.06.20050575v1https://www.medrxiv.org/content/10.1101/2020.04.03.20051722v1https://www.ncbi.nlm.nih.gov/pubmed/25705421https://www.kegg.jp/kegg-bin/show_pathway?map04610https://www.ncbi.nlm.nih.gov/pubmed/25544351https://www.ncbi.nlm.nih.gov/pubmed/26670422https://www.ncbi.nlm.nih.gov/pubmed/26670422https://www.ncbi.nlm.nih.gov/pubmed/12371961https://www.ncbi.nlm.nih.gov/pubmed/12371961https://www.ncbi.nlm.nih.gov/pubmed/30283040https://www.ncbi.nlm.nih.gov/pubmed/25482567https://www.ncbi.nlm.nih.gov/pubmed/25482567https://www.ncbi.nlm.nih.gov/pubmed/18544849https://www.ncbi.nlm.nih.gov/pubmed/30767945https://www.ncbi.nlm.nih.gov/pubmed/30767945

  • 45 Mechanisms of severe acute respiratory syndrome coronavirus-induced acute lung injury. Gralinski LE, Bankhead A 3rd, Jeng S, Menachery VD, Proll S, Belisle SE, Matzke M, Webb-Robertson BJ, Luna ML, Shukla AK, Ferris MT, Bolles M, Chang J, Aicher L, Waters KM, Smith RD, Metz TO, Law GL, Katze MG, McWeeney S, Baric RS. mBio. 2013 Aug 6;4(4). pii: e00271-13. doi: 10.1128/mBio.00271-13. PMID:23919993

    46 https://www.genome.jp/kegg-bin/show_pathway?map04610

    47 IL-1 beta induces urokinase-plasminogen activator expression and cell migration through PKC alpha, JNK1/2, and NF-kappaB in A549 cells. Cheng CY, Hsieh HL, Sun CC, Lin CC, Luo SF, Yang CM. J Cell Physiol. 2009 Apr;219(1):183-93. doi: 10.1002/jcp.21669. PMID:19097143

    48 Tumor necrosis factor α stimulates expression and secretion of urokinase plasminogen activator in human dental pulp cells. Narita T, Muromachi K, Kamio N, Nakao S, Matsushima K, Hashizume H. J Oral Sci. 2012;54(4):329-36. PMID:23221158

    49 Bacterial endotoxin enhances colorectal cancer cell adhesion and invasion through TLR-4 and NF-kappaB-dependent activation of the urokinase plasminogen activator system. Killeen SD, Wang JH, Andrews EJ, Redmond HP. Br J Cancer. 2009 May 19;100(10):1589-602. doi: 10.1038/sj.bjc.6604942. PMID:19436306

    50 Rat urinary kallikrein. Chao J, Chao L. Methods Enzymol. 1988;163:128-43. PMID:3070295

    51 https://www.genome.jp/kegg-bin/show_pathway?hsa04610

    52 Hypokalemia and Clinical Implications in Patients with Coronavirus Disease 2019 (COVID-19), https://www.medrxiv.org/content/10.1101/2020.02.27.20028530v1

    53 Influences of up-regulation of miR-126 on septic inflammation and prognosis through AKT/Rac1 signaling pathway. Wang HF, Wang YQ, Dou L, Gao HM, Wang B, Luo N, Li Y. Eur Rev Med Pharmacol Sci. 2019 Mar;23(5):2132-2138. doi: 10.26355/eurrev_201903_17257. PMID:30915758

    54 Serum miR-126-3p level is down-regulated in sepsis patients. Chen C, Zhang L, Huang H, Liu S, Liang Y, Xu L, Li S, Cheng Y, Tang W. Int J Clin Exp Pathol. 2018 May 1;11(5):2605-2612. PMID:31938374

    55 Protective Effects of MicroRNA-126 on Human Cardiac Microvascular Endothelial Cells Against Hypoxia/Reoxygenation-Induced Injury and Inflammatory Response by Activating PI3K/Akt/eNOS Signaling Pathway. Yang HH, Chen Y, Gao CY, Cui ZT, Yao JM. Cell Physiol Biochem. 2017;42(2):506-518. doi: 10.1159/000477597. PMID:28578351

    56 Cellular and molecular mechanisms of pomegranate juice-induced anti-metastatic effect on prostate cancer cells. Wang L, Alcon A, Yuan H, Ho J, Li QJ, Martins-Green M. Integr Biol (Camb). 2011 Jul;3(7):742-54. doi: 10.1039/c0ib00122h. PMID:21594291

    57 Pomegranate polyphenolics suppressed azoxymethane-induced colorectal aberrant crypt foci and inflammation: possible role of miR-126/VCAM-1 and miR-126/PI3K/AKT/mTOR. Banerjee N, Kim H, Talcott S, Mertens-Talcott S. Carcinogenesis. 2013 Dec;34(12):2814-22. doi: 10.1093/carcin/bgt295. PMID:23996930

    https://www.ncbi.nlm.nih.gov/pubmed/23919993https://www.genome.jp/kegg-bin/show_pathway?map04610https://www.ncbi.nlm.nih.gov/pubmed/19097143https://www.ncbi.nlm.nih.gov/pubmed/19097143https://www.ncbi.nlm.nih.gov/pubmed/23221158https://www.ncbi.nlm.nih.gov/pubmed/23221158https://www.ncbi.nlm.nih.gov/pubmed/19436306https://www.ncbi.nlm.nih.gov/pubmed/19436306https://www.ncbi.nlm.nih.gov/pubmed/3070295https://www.genome.jp/kegg-bin/show_pathway?hsa04610https://www.medrxiv.org/content/10.1101/2020.02.27.20028530v1https://www.ncbi.nlm.nih.gov/pubmed/30915758https://www.ncbi.nlm.nih.gov/pubmed/30915758https://www.ncbi.nlm.nih.gov/pubmed/31938374https://www.ncbi.nlm.nih.gov/pubmed/28578351https://www.ncbi.nlm.nih.gov/pubmed/28578351https://www.ncbi.nlm.nih.gov/pubmed/28578351https://www.ncbi.nlm.nih.gov/pubmed/21594291https://www.ncbi.nlm.nih.gov/pubmed/21594291https://www.ncbi.nlm.nih.gov/pubmed/23996930https://www.ncbi.nlm.nih.gov/pubmed/23996930

  • 58 http://mirtarbase.cuhk.edu.cn/php/search.php?opt=search_box&kw=HMGB1&sort=sum_evidences& order=desc

    59 The Influence of Diet on MicroRNAs that Impact Cardiovascular Disease. Kura B, Parikh M, Slezak J, Pierce GN. Molecules. 2019 Apr 17;24(8). pii: E1509. doi: 10.3390/molecules24081509. PMID:30999630

    60 MicroRNA-Mediated Regulation of HMGB1 in Human Hepatocellular Carcinoma. Yan J, Ying S, Cai X. Biomed Res Int. 2018 Jan 31;2018:2754941. doi: 10.1155/2018/2754941. PMID:29651425

    61 MicroRNA-548 regulates high mobility group box 1 expression in patients with preterm birth and chorioamnionitis. Son GH, Kim Y, Lee JJ, Lee KY, Ham H, Song JE, Park ST, Kim YH. Sci Rep. 2019 Dec 24;9(1):19746. doi: 10.1038/s41598-019-56327-9. PMID:31875024

    62 Effect of microRNA-129-5p targeting HMGB1-RAGE signaling pathway on revascularization in a collagenase-induced intracerebral hemorrhage rat model. Ma XL, Li SY, Shang F. Biomed Pharmacother. 2017 Sep;93:238-244. doi: 10.1016/j.biopha.2017.06.012. PMID:28645008

    63 Elevated microRNA-129-5p level ameliorates neuroinflammation and blood-spinal cord barrier damage after ischemia-reperfusion by inhibiting HMGB1 and the TLR3-cytokine pathway. Li XQ, Chen FS, Tan WF, Fang B, Zhang ZL, Ma H. J Neuroinflammation. 2017 Oct 23;14(1):205. doi: 10.1186/s12974-017-0977-4. PMID:29061187

    64 MicroRNA-34a directly targets high-mobility group box 1 and inhibits the cancer cell proliferation, migration and invasion in cutaneous squamous cell carcinoma. Li S, Luo C, Zhou J, Zhang Y. Exp Ther Med. 2017 Dec;14(6):5611-5618. doi: 10.3892/etm.2017.5245. PMID:29285100

    65 https://cancerres.aacrjournals.org/content/canres/early/2015/02/26/0008-5472.CAN-14-2147.full.pdf

    66 miR-193a-3p interaction with HMGB1 downregulates human endothelial cell proliferation and migration. Khoo CP, Roubelakis MG, Schrader JB, Tsaknakis G, Konietzny R, Kessler B, Harris AL, Watt SM. Sci Rep. 2017 Mar 9;7:44137. doi: 10.1038/srep44137. PMID:28276476

    67 The Role of Bioactive Dietary Components in Modulating miRNA Expression in Colorectal Cancer. Gavrilas LI, Ionescu C, Tudoran O, Lisencu C, Balacescu O, Miere D. Nutrients. 2016 Sep 26;8(10). pii: E590. PMID:27681738

    68 Nutrimiromics: Role of microRNAs and Nutrition in Modulating Inflammation and Chronic Diseases. Quintanilha BJ, Reis BZ, Duarte GBS, Cozzolino SMF, Rogero MM. Nutrients. 2017 Oct 27;9(11). pii: E1168. doi: 10.3390/nu9111168. PMID:29077020

    69 Quercetin, Epigallocatechin Gallate, Curcumin, and Resveratrol: From Dietary Sources to Human MicroRNA Modulation. Cione E, La Torre C, Cannataro R, Caroleo MC, Plastina P, Gallelli L. Molecules. 2019 Dec 23;25(1). pii: E63. doi: 10.3390/molecules25010063. PMID:31878082

    70 Cellular and molecular mechanisms of pomegranate juice-induced anti-metastatic effect on prostate cancer cells. Wang L, Alcon A, Yuan H, Ho J, Li QJ, Martins-Green M. Integr Biol (Camb). 2011 Jul;3(7):742-54. doi: 10.1039/c0ib00122h. PMID:21594291

    http://mirtarbase.cuhk.edu.cn/php/search.php?opt=search_box&kw=HMGB1&sort=sum_evidences&%20order=deschttp://mirtarbase.cuhk.edu.cn/php/search.php?opt=search_box&kw=HMGB1&sort=sum_evidences&%20order=deschttps://www.ncbi.nlm.nih.gov/pubmed/30999630https://www.ncbi.nlm.nih.gov/pubmed/29651425https://www.ncbi.nlm.nih.gov/pubmed/31875024https://www.ncbi.nlm.nih.gov/pubmed/31875024https://www.ncbi.nlm.nih.gov/pubmed/28645008https://www.ncbi.nlm.nih.gov/pubmed/28645008https://www.ncbi.nlm.nih.gov/pubmed/29061187https://www.ncbi.nlm.nih.gov/pubmed/29061187https://www.ncbi.nlm.nih.gov/pubmed/29285100https://www.ncbi.nlm.nih.gov/pubmed/29285100https://cancerres.aacrjournals.org/content/canres/early/2015/02/26/0008-5472.CAN-14-2147.full.pdfhttps://www.ncbi.nlm.nih.gov/pubmed/28276476https://www.ncbi.nlm.nih.gov/pubmed/27681738https://www.ncbi.nlm.nih.gov/pubmed/29077020https://www.ncbi.nlm.nih.gov/pubmed/31878082https://www.ncbi.nlm.nih.gov/pubmed/31878082https://www.ncbi.nlm.nih.gov/pubmed/21594291https://www.ncbi.nlm.nih.gov/pubmed/21594291

  • 71 Mango polyphenolics suppressed tumor growth in breast cancer xenografts in mice: role of the PI3K/AKT pathway and associated microRNAs. Banerjee N, Kim H, Krenek K, Talcott ST, Mertens-Talcott SU. Nutr Res. 2015 Aug;35(8):744-51. PMID:26194618 72 Mango polyphenolics reduce inflammation in intestinal colitis-involvement of the miR-126/PI3K/AKT/mTOR axis in vitro and in vivo. Kim H, Banerjee N, Barnes RC, Pfent CM, Talcott ST, Dashwood RH, Mertens-Talcott SU. Mol Carcinog. 2017 Jan;56(1):197-207. PMID:27061150

    73 Profiles of Circulating MiRNAs Following Metformin Treatment in Patients with Type 2 Diabetes. Demirsoy İH, Ertural DY, Balci Ş, Çınkır Ü, Sezer K, Tamer L, Aras N. J Med Biochem. 2018 Dec 1;37(4):499-506. doi: 10.2478/jomb-2018-0009. PMID:30584410

    74 Resveratrol protects mice against SEB-induced acute lung injury and mortality by miR-193a modulation that targets TGF-β signalling. Alghetaa H, Mohammed A, Sultan M, Busbee P, Murphy A, Chatterjee S, Nagarkatti M, Nagarkatti P. J Cell Mol Med. 2018 May;22(5):2644-2655. doi: 10.1111/jcmm.13542. PMID:29512867

    75 Therapeutic potential of an anti-diabetic drug, metformin: alteration of miRNA expression in prostate cancer cells. Avci CB, Harman E, Dodurga Y, Susluer SY, Gunduz C. Asian Pac J Cancer Prev. 013;14(2):765-8.PMID:23621234

    76 Promiscuous Effects of Some Phenolic Natural Products on Inflammation at Least in Part Arise from Their Ability to Modulate the Expression of Global Regulators, Namely microRNAs. Tili E, Michaille JJ. Molecules. 2016 Sep 21;21(9). pii: E1263. PMID:27657035

    77 Activation of the SIRT1/p66shc antiapoptosis pathway via carnosic acid-induced inhibition of miR-34a protects rats against nonalcoholic fatty liver disease. Shan W, Gao L, Zeng W, Hu Y, Wang G, Li M, Zhou J, Ma X, Tian X, Yao J. Cell Death Dis. 2015 Jul 23;6:e1833. doi: 10.1038/cddis.2015.196. PMID:26203862

    78 Proton pump inhibitors (PPIs) impact on tumour cell survival, metastatic potential and chemotherapy resistance, and affect expression of resistance-relevant miRNAs in esophageal cancer. Lindner K, Borchardt C, Schöpp M, Bürgers A, Stock C, Hussey DJ, Haier J, Hummel R. J Exp Clin Cancer Res. 2014 Sep 1;33:73. doi: 10.1186/s13046-014-0073-x. PMID:25175076

    79 Cross-Regulation of Proinflammatory Cytokines by Interleukin-10 and miR-155 in Orientia tsutsugamushi-Infected Human Macrophages Prevents Cytokine Storm. Tsai MH, Chang CH, Tsai RK, Hong YR, Chuang TH, Fan KT, Peng CW, Wu CY, Hsu WL, Wang LS, Chen LK, Yu HS. J Invest Dermatol. 2016 Jul;136(7):1398-1407. doi: 10.1016/j.jid.2015.11.034. PMID:26921773

    80 Dietary apigenin reduces LPS-induced expression of miR-155 restoring immune balance during inflammation. Arango D, Diosa-Toro M, Rojas-Hernandez LS, Cooperstone JL, Schwartz SJ, Mo X, Jiang J, Schmittgen TD, Doseff AI. Mol Nutr Food Res. 2015 Apr;59(4):763-72. doi: 10.1002/mnfr.201400705. PMID:25641956

    81 Potential miRNA involvement in the anti-adipogenic effect of resveratrol and its metabolites. Eseberri I, Lasa A, Miranda J, Gracia A, Portillo MP. PLoS One. 2017 Sep 27;12(9):e0184875. doi: 10.1371/journal.pone.0184875. PMID:28953910

    https://www.ncbi.nlm.nih.gov/pubmed/26194618https://www.ncbi.nlm.nih.gov/pubmed/26194618https://www.ncbi.nlm.nih.gov/pubmed/27061150https://www.ncbi.nlm.nih.gov/pubmed/27061150https://www.ncbi.nlm.nih.gov/pubmed/30584410https://www.ncbi.nlm.nih.gov/pubmed/29512867https://www.ncbi.nlm.nih.gov/pubmed/29512867https://www.ncbi.nlm.nih.gov/pubmed/23621234https://www.ncbi.nlm.nih.gov/pubmed/23621234https://www.ncbi.nlm.nih.gov/pubmed/27657035https://www.ncbi.nlm.nih.gov/pubmed/27657035https://www.ncbi.nlm.nih.gov/pubmed/26203862https://www.ncbi.nlm.nih.gov/pubmed/26203862https://www.ncbi.nlm.nih.gov/pubmed/25175076https://www.ncbi.nlm.nih.gov/pubmed/25175076https://www.ncbi.nlm.nih.gov/pubmed/26921773https://www.ncbi.nlm.nih.gov/pubmed/26921773https://www.ncbi.nlm.nih.gov/pubmed/25641956https://www.ncbi.nlm.nih.gov/pubmed/25641956https://www.ncbi.nlm.nih.gov/pubmed/28953910

  • 82 Effect of quercetin and its metabolites isorhamnetin and quercetin-3-glucuronide on inflammatory gene expression: role of miR-155. Boesch-Saadatmandi C, Loboda A, Wagner AE, Stachurska A, Jozkowicz A, Dulak J, Döring F, Wolffram S, Rimbach G. J Nutr Biochem. 2011 Mar;22(3):293-9. doi: 10.1016/j.jnutbio.2010.02.008. PMID:20579867

    83 The Influence of Diet on MicroRNAs that Impact Cardiovascular Disease. Kura B, Parikh M, Slezak J, Pierce GN. Molecules. 2019 Apr 17;24(8). pii: E1509. doi: 10.3390/molecules24081509. PMID:30999630

    84 https://chemrxiv.org/articles/Homology_Modeling_and_Docking_Studies_of_TMPRSS2_with_Experimentally_ Known_Inhibitors_Camostat_Mesylate_Nafamostat_and_Bromhexine_Hydrochloride_to_Control_SARS-Coronavirus-2/12162360

    85 Probing the substrate specificities of matriptase, matriptase-2, hepsin and DESC1 with internally quenched

    fluorescent peptides. Béliveau F, Désilets A, Leduc R. FEBS J. 2009 Apr;276(8):2213-26. doi:

    10.1111/j.1742-4658.2009.06950.x. PMID:9302215

    86 Heparin sensitivity test for patients requiring cardiopulmonary bypass. DeBois WJ, Liu J, Elmer B,

    Ebrahimi H, Voevidko L, Lee LY, Krieger KH, Isom WW, Girardi LN. J Extra Corpor Technol. 2006

    Dec;38(4):307-9. PMID:17312901

    87 Plant-derived protein tyrosine kinase inhibitors as anticancer agents. Hollósy F, Kéri G. Curr Med Chem Anticancer Agents. 2004 Mar;4(2):173-97. PMID:15032721

    88 Dietary Apigenin Reduces Induction of LOX-1 and NLRP3 Expression, Leukocyte Adhesion, and

    Acetylated Low-Density Lipoprotein Uptake in Human Endothelial Cells Exposed to Trimethylamine-N-

    Oxide. Yamagata K, Hashiguchi K, Yamamoto H, Tagami M. J Cardiovasc Pharmacol. 2019 Dec;74(6):558-

    565. doi: 10.1097/FJC.0000000000000747. PMID:31815868

    Flavonoids interfere with NLRP3 inflammasome activation. Lim H, Min DS, Park H, Kim HP. Toxicol Appl

    Pharmacol. 2018 Sep 15;355:93-102. doi: 10.1016/j.taap.2018.06.022. PMID:29960001 (Specifically

    apigenin)

    https://www.ncbi.nlm.nih.gov/pubmed/20579867https://www.ncbi.nlm.nih.gov/pubmed/20579867https://www.ncbi.nlm.nih.gov/pubmed/30999630https://chemrxiv.org/articles/Homology_Modeling_and_Docking_Studies_of_TMPRSS2_with_Experimentally_%20Known_Inhibitors_Camostat_Mesylate_Nafamostat_and_Bromhexine_Hydrochloride_to_Control_SARS-Coronavirus-2/12162360https://chemrxiv.org/articles/Homology_Modeling_and_Docking_Studies_of_TMPRSS2_with_Experimentally_%20Known_Inhibitors_Camostat_Mesylate_Nafamostat_and_Bromhexine_Hydrochloride_to_Control_SARS-Coronavirus-2/12162360https://chemrxiv.org/articles/Homology_Modeling_and_Docking_Studies_of_TMPRSS2_with_Experimentally_%20Known_Inhibitors_Camostat_Mesylate_Nafamostat_and_Bromhexine_Hydrochloride_to_Control_SARS-Coronavirus-2/12162360https://www.ncbi.nlm.nih.gov/pubmed/19302215https://www.ncbi.nlm.nih.gov/pubmed/19302215https://www.ncbi.nlm.nih.gov/pubmed/17312901https://www.ncbi.nlm.nih.gov/pubmed/15032721https://www.ncbi.nlm.nih.gov/pubmed/31815868https://www.ncbi.nlm.nih.gov/pubmed/31815868https://www.ncbi.nlm.nih.gov/pubmed/31815868https://www.ncbi.nlm.nih.gov/pubmed/29960001