Loading

Review Article Open Access
Volume 4 | Issue 4 | DOI: https://doi.org/10.33696/Signaling.4.106

Retinoic Acid Induced Cell Signaling as a Counter Against Disease

  • 1Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
+ Affiliations - Affiliations

*Corresponding Author

Justin Franco, Justin.Franco@rockets.utoledo.edu

Received Date: September 21, 2023

Accepted Date: November 06, 2023

Abstract

Many disease processes result from disruption of physiologic cell signaling pathways. Cancer often develops from the loss of cell cycle regulation, while inflammatory disease results from dysregulated immune activity. Likewise, many microbial infections avoid immune clearance by interfering with cellular antimicrobial pathways. Retinoic Acid (RA) is a dynamic compound, derived from vitamin A, that can regulate various signaling pathways. RA induced cell signaling has proven beneficial against different diseases, such as Acute Promyelocytic Leukemia (APL) and psoriasis. Against APL, RA induces cellular differentiation in cancer cells to restore proper function. In psoriasis, RA downregulates inflammatory pathways, such as NF-κB. RA’s anti-inflammatory properties have also been examined in the context of sepsis, where recent animal studies have shown positive benefits. Along with regulating inflammation, RA exhibits indirect antimicrobial properties. Unlike conventional antimicrobials which target pathogens directly, RA functions as a host-directed therapy (HDT), promoting cell antimicrobial defenses. Recent studies examining RA have shown that it can improve macrophage clearance of microbial pathogens and stimulate the antiviral type-I interferon (IFN) response. RA’s effectiveness has been demonstrated against clinically relevant pathogens, such as Mycobacterium tuberculosis, Aspergillus fumigatus, and Measles virus. In this review, the therapeutic potential of RA to treat various diseases by regulating cell signaling pathways will be explored.

Keywords

Retinoic Acid, Anti-inflammatory, Cell Signaling Pathways, Interferon Signaling

Abbreviations

ADH: Alcohol Dehydrogenase; AMP: Antimicrobial Protein; APL: Acute Promyelocytic Leukemia; ATRA: All-trans Retinoic Acid; BC01: β-carotene-15, 15'-dioxygenase; CEACAM1: Carcinoembryonic Antigen-related Cell Adhesion Molecule 1; CRABP: Cellular Retinoic Acid Binding Protein; HDT: Host-directed Therapy; IFIT: Interferon-induced Tetratricopeptide repeat protein; IFIT: Interferon-inducible Transmembrane protein; IFN: Interferon; IL: Interleukin; IRF: Interferon Regulatory Factor; MAPK: Mitogen Activated Protein Kinase; MDA5: Melanoma Differentiation Associated gene 5; MxA: Myxovirus resistance protein 1; NF-κB: Nuclear Factor κ B; PGE-2: Prostaglandin E-2; PML: Promyelocytic Leukemia protein; PRR: Pattern Recognition Receptor; RA: Retinoic Acid; RALDH: Retinal Dehydrogenase; RAR: Retinoic Acid Receptor; RARE: Retinoic Acid Response Element; RBP: Retinol Binding Protein; RIG-I: Retinoic Acid Inducible Gene I; RXR: Retinoid X Receptor; SARS-COV-2: Severe Acute Respiratory Syndrome Coronavirus 2; TBK-1: TANK-binding Kinase 1; TNF-α: Tumor Necrosis Factor α

Introduction to Retinoic Acid

Vitamins are essential nutrients that are required for biological processes, but are unable to be synthesized by the human body in large amounts [1]. In general, vitamins are classified into two groups based on their chemical properties: water-soluble (e.g., vitamin B1, B2, B3, etc.) and fat-soluble (e.g., vitamin A, C, D, etc.) [1]. Vitamin A is a critical fat-soluble nutrient that plays a key role in signaling pathways involved in fetal development, tissue differentiation, and immune regulation [2-4]. It is also a major component in the visual system, where it interacts with opsin proteins to induce visual activity [5]. Vitamin A’s effect on cellular signaling pathways results from its function as a transcription factor for different genes [2-4]. For example, vitamin A regulates fetal development by promoting the expression of homeobox genes, which are responsible for embryogenesis [4]. Because of its extensive physiological activity, vitamin A deficiency results in widespread symptoms [6]. Common symptoms of vitamin A deficiency include night blindness, mucosal membrane breakdown, and increased risk of infection (i.e., immune dysfunction) [6].

Like other essential nutrients, vitamin A can’t be synthesized and must be acquired from the diet [3, 7]. Fortunately, vitamin A is present in multiple dietary sources, such as animal (e.g., meat, milk, dairy products, eggs, etc.) and plant products (e.g., carrots, spinach, tomatoes, oranges, etc.) [7]. Unlike animal products which contain preformed vitamin A (Retinol), plant products produce provitamin A carotenoids, such as β-carotene [7,8]. The human body is able to make use of both Retinol and provitamin A carotenoids by metabolizing them into retinoic acid (RA) for cellular use [8].

The conversion of vitamin A into its active form (i.e., RA) occurs in a stepwise process (Figure 1). First, retinol and β-carotene are absorbed by intestinal epithelial cells into circulation [7,9]. In serum, Retinol and β-carotene are transported via retinol binding protein (RBP) and chylomicrons, respectively [3,8]. Circulating retinol and β-carotene are then taken up by liver cells, which contain the highest concentration of vitamin A in the human body [3,7]. Within liver cells, retinol is converted to retinal by alcohol dehydrogenase (ADH) [3,7,10]. β-carotene is also converted to retinal, via the activity of β-carotene-15,15′-dioxygenase (BCO1) [8]. Lastly, retinal gets metabolized into RA by retinal dehydrogenase (RALDH) [3,7,10]. The primary forms of RA include all-trans RA (ATRA), 9-cis RA, and 13-cis RA [3,9]. RA binds to cellular RA binding protein (CRABP) and is transported into the nucleus, where it activates RA specific transcription factors [3,7,10]. Of these three metabolites, ATRA demonstrates the greatest transcriptional activity, capable of binding to RA receptors (RARs) within the nucleus [11,12]. 9-cis RA predominantly binds to retinoid X receptors (RXRs) however, it can also interact with RARs [11,12]. The binding activity of 13-cis RA is unknown [11,12].

Figure 1. Summary of vitamin A metabolism. Retinol and β-carotene get taken up by the cell and metabolized into RA, the active form of vitamin A. RA is then transported into the nucleus by CRABP, where it activates the nuclear transcription factors RAR and RXR. Activation of RAR and RXR leads to RARE gene expression. Created with BioRender.com.

Both RAR and RXRs consist of three isoforms: α, β, and γ [12]. RAR is typically bound to RXR as a heterodimer (i.e., RAR/RXR), while RXR is usually present as an inactive tetramer [11-13]. RAR/RXR is bound to RA response elements (RAREs) in the genome [12]. Without RA present, RAR/RXR represses gene expression by recruiting corepressors that promote histone deacetylase complexes [11,12]. With RA present, RAR/RXR induces gene expression by recruiting coactivators that promote histone acetyl transferase activity [11,12]. RA induced RAR/RXR activation results in the expression of RAREs, which encompass numerous genes involved in different signaling pathways (Figure 1) [11,12,14]. For instance, RAREs include genes that regulate fetal development (e.g., Hoxa1, Hoxa4, etc.), immune function (e.g., IL2RA, Socs3, Tgfbr1 etc.), cell growth (e.g., MYC, MYCN, etc.), and RA activity (e.g., RARA, RARB, etc.) (Table 1) [14, 15]. The transcriptional activity of RXR is less understood than that of RAR/RXR. However, previous studies have shown that RA induces RXR binding to other transcription factors to promote their activity [13,16]. Noticeable transcription factors that are promoted by RXR include thyroid hormone receptor and peroxisome proliferator-activated receptor [13]. Because the effect of RA on RARE expression is more clearly established, the review will focus on the activity of RAR/RXR.

Table 1. Listed RARE Gene Activity.

Gene

Full Name

Function

Hoxa1

Homeobox A1

Regulates cellular differentiation

Hoxa4

Homeobox A4

IL2RA

Interleukin 2 Receptor subunit α

Forms interleukin 2 receptor, a key mediator of inflammation

MYC

MYC proto-oncogene

Regulates cell cycle progression and apoptosis

MYCN

MYCN proto-oncogene

RARA

Retinoic Acid Receptor α

Regulates expression of RAREs

RARB

Retinoic Acid Receptor β

Socs3

Suppressor of Cytokine Signaling 3

Promotes anti-inflammatory activity

Tgfbr1

Transforming Growth Factor β Receptor 1

Forms Transforming Growth Factor β receptor, which promotes anti-inflammatory activity

ATRA Induced Cell Signaling as a Disease Treatment

Therapeutic rationale

Due to its effect on different cellular signaling pathways, ATRA has been extensively studied as a potential therapeutic for diseases resulting from dysregulated signaling pathways. In oncology, ATRA is predominantly used to treat acute promyelocytic leukemia (APL), where it promotes cellular differentiation and induces apoptosis in cancer cells [11,17-19]. During APL, promyelocytes have lost the ability to differentiate into mature granulocytes, so they overcrowd the bone marrow and disrupt other blood cells (e.g., platelets, red blood cells, etc.) [17-19]. Because of its effectiveness in treating APL, ATRA is listed by the World Health Organization as an essential medication for healthcare systems [20]. RA derivatives have also shown positive benefits in the context of melanoma, hepatoma, lung, breast, and prostate cancer [21,22]. In dermatology, ATRA is used to treat severe forms of acne and inflammatory skin conditions, such as psoriasis [11,23]. ATRA can reduce inflammation by upregulating anti-inflammatory signaling in immune cells [3,11,23]. Its anti-inflammatory properties have also been studied in the context of sepsis, where it can downregulate proinflammatory signaling pathways (i.e., NF-κB pathway) [3,24-26].

Although ATRA has predominantly been studied in the context of cancer and inflammatory disease, recent studies have also highlighted its impact on innate immune pathways. Unlike conventional antimicrobials that directly target their associated pathogen, ATRA can enhance immune pathways to improve bacterial and viral clearance [3,27]. This therapeutic strategy, known as host-directed therapy (HDT), has gained interest as an additional treatment option, especially against antibiotic resistant bacteria [28,29]. Resulting in approximately 1.27 million deaths globally in 2019, antibiotic resistance is a significant health problem requiring renewed focus and novel therapeutics [30,31]. Likewise, the emergence of novel viral pathogens necessitates an expansion of antiviral treatments [32].

ATRA induction of differentiation in cancer

Cancer is a multifaceted disease process, encompassing different pathologies that can affect every organ system. Although the causes of cancer are numerous, cancer cells generally exhibit dysregulated cell signaling that results in excessive proliferation, loss of differentiation, and metastasis [33]. Loss of regular signaling pathways often occurs due to mutations in proto-oncogenes or tumor suppressor genes [33-35]. Proto-oncogenes regulate cell growth, so when they become mutated (i.e., oncogenes) they can induce abnormal proliferation [34]. Tumor suppressor genes maintain normal cellular signaling by regulating cell cycle progression and DNA repair pathways [35].

APL is a blood cancer caused by abnormal promyelocytes [36,37]. During erythropoiesis, promyelocytes develop in bone marrow and differentiate into granulocytes (e.g., neutrophils, eosinophils, basophils) [38,39]. In APL, promyelocytes are unable to differentiate, and instead proliferate and crowd out normal developing blood cells (e.g., platelets, red blood cells, etc.) [36,37]. Clinically, this manifests as abnormal white blood cell counts (i.e., promyelocyte proliferation), decreased platelets, and bleeding [36]. Without treatment APL is a fatal disease, with death resulting from hemorrhage due to reduced platelet levels [36]. ATRA treatment has revolutionized APL care, producing response rates of 90% [36,40]. Unfortunately, the high response rate of ATRA monotherapy is tempered by its low remission rate, with 20-30% of patients experiencing APL recurrence [36]. To combat high recurrence rates, current APL treatments utilize combination therapies, relying on ATRA plus other chemotherapy agents (e.g., arsenic trioxide, idarubicin, etc.). Combination therapies drastically improve APL survival by demonstrating complete remission and 5 year survival rates of 91% and 95%, respectively [36].

ATRA treats APL by restoring cell signaling, thus enabling proper gene expression and cellular differentiation. The loss of cellular differentiation in APL is due to a balanced translocation between chromosomes 15 and 17 [36,37]. This results in the formation of a fusion protein involving Promyelocytic Leukemia protein and RARα (PML-RARα) [36,37]. Under physiologic conditions, PML functions as a tumor suppressor protein, regulating cell cycle progression and DNA repair [41,42]. However, when fused to RARα it disrupts normal gene expression by recruiting corepressors, histone modifying proteins, and by blocking RARE [37,43]. ATRA interferes with PML-RARα by directly binding to RARα [43]. After binding ATRA, PML-RARα undergoes conformational changes that prevent it from blocking RAREs or recruiting corepressors and histone modifying proteins [43]. Without interference from PML-RARα, normal cell signaling is restored and the promyelocyte can complete the differentiation process (Figure 2).

Figure 2. ATRA induced differentiation in APL. ATRA binding to PML-RARα allows for RARE gene expression, preventing cancer induced recruitment of gene silencing proteins (e.g., Corepressors and histone modifying proteins). Created with BioRender.com.

The effectiveness of ATRA’s antineoplastic properties during APL has led to increased study. Similar to ATRA, RA derivatives are also effective against different blood cancers. For example, Bexarotene is an RA derivative approved by US FDA for the treatment of cutaneous T cell lymphoma [7,22,44]. Positive findings have also been encountered by in vitro studies, where RA derivatives were shown to promote cellular differentiation and disrupt proliferation in various cancer cell-lines (e.g., lung, breast, etc.) [7,22,45]. However, these studies have been unable to translate into the clinic, with experiments failing to demonstrate a survival benefit [22,45]. The discrepancy between in vitro and clinical studies demonstrates the difficulty of modeling cancer, which can recruit noncancerous cells and adapt to monotherapies. Further study examining ATRA in combination with other therapies (e.g., chemotherapy, immunotherapy, etc.) may better elucidate its effectiveness against cancer.

RA has exhibited successful use as a chemo-preventative agent [46]. Unlike chemotherapy agents that directly induce cancer cell death, chemo-preventative agents disrupt carcinogenesis to halt cancer formation [46]. As a chemo-preventative agent, ATRA can reduce cancer progression of oral leukoplakia, which is a potentially malignant disorder [47]. Likewise, 13-cis RA can reduce tumor formation in patients with xeroderma pigmentosum, who exhibit increased rates of non-melanoma skin cancer due to nonfunctional DNA repair proteins [48]. In this context, ATRA’s effect is explained by its capacity to upregulate tumor suppressor genes, such as p53, and facilitate activation of apoptotic pathways [49,50]. ATRA’s antineoplastic properties have been applied with mixed results, necessitating further research to determine the conditions most susceptible to treatment.

ATRA downregulation of inflammatory disease processes

Inflammatory disease is a complex pathologic process with diverse clinical presentations. Depending on the degree of inflammation, inflammatory disease can affect a single organ system (e.g., psoriasis, atherosclerosis, etc.) or act systemically, such as in sepsis [51]. Regardless of severity, inflammatory processes are mediated by the release of proinflammatory cytokines (e.g., TNFα, IL-6, etc.) [51,52]. These cytokines are produced in response to activation of inflammatory signaling pathways, such as the NF-κB or MAPK pathway [51-54]. During inflammatory disease, these pathways become dysregulated and result in excessive inflammation.

An inflammatory disease characterized by chronic inflammation in the skin is psoriasis [55]. Psoriasis presents clinically with scaly red plaques that are defined by epidermal proliferation and immune cell infiltration [55-57]. Treatments for psoriasis include anti-inflammatory therapeutics (e.g., infliximab, utekinumab, etc.) and retinoids (e.g., acitretin, tazarotene, etc.) [56,57]. Retinoids improve psoriasis symptoms by reducing epidermal proliferation and downregulating inflammatory cytokines [56,57]. Specifically, retinoid induced activation of RARE leads to decreased IL-6 expression [57].

Another chronic inflammatory condition is atherosclerosis, which is defined by cholesterol buildup that induces inflammation in the vessel wall [58]. This inflammation results in vascular dysfunction that can lead to clot formation, thus disrupting blood flow to downstream organs [58]. Unlike psoriasis, atherosclerosis treatment focuses on limiting cholesterol accumulation and reducing clotting risk. ATRA’s efficacy in treating atherosclerosis is currently under investigation [59]. However, its anti-inflammatory effect on mediators of atherosclerosis, such as macrophages and Tregs, highlights a possibility to reduce disease progression [58,59]. RA’s anti-inflammatory effect has been extensively studied, with in vitro studies illustrating its cell type dependent activity. In macrophages RA decreases TNFα, IL-12, and PGE-2 secretion by interfering with NF-κB activity [25,26,60,61]. This interference has been proposed to occur via direct binding by RXR [26,61] (Figure 3). ATRA also stimulates Treg cell expansion, which is a major source of the anti-inflammatory cytokine IL-10 [62].

Figure 3. ATRA’s anti-inflammatory effect. ATRA reduces inflammation by upregulating anti-inflammatory genes and disrupting NF-κB activity. Created with BioRender.com.

Because of its widespread influence on immune function, RA has been examined as a potential treatment for sepsis [26,63]. Sepsis is a fatal disease characterized as a dysfunctional host response towards an infection with risk of organ failure [63-65]. Regardless of etiology, many of the symptoms associated with sepsis (e.g., fever, excessive clotting, hypotension) result from excessive inflammation [63]. These inflammatory processes are mediated by a dysregulated release of proinflammatory cytokines, such as TNFα, IL-6, and IL-8 [63,66,67]. Current treatments with antimicrobials and corticosteroids (i.e., anti-inflammatory agent) have improved survival rates from sepsis [65,68]. However, mortality rates still range from 15 to 20%, and can increase to between 20 and 50% in cases of septic shock [66,69]. Its severe clinical presentation leads to over 5 million deaths globally [67].

With mortality rates from sepsis still high, ATRA has been examined as a potential treatment option. In lipopolysaccharide induced sepsis animal models, ATRA downregulates NF-κB activation and increases survival rates [24,26]. Similar results were seen in models of polymicrobial sepsis, where ATRA increased survival and was associated with decreased proinflammatory cytokine expression [26]. Clinical studies are needed to confirm RA’s potential benefit during sepsis; however, previous work supports a promising outlook for its use.

ATRA activation of antimicrobial defense

Under typical conditions, the immune system is effective at clearing microbial pathogens. Activation of the innate immune system is facilitated by cellular pattern recognition receptors (PRRs) that recognize invading pathogens and trigger an inflammatory response [63,70]. The release of proinflammatory cytokines leads to leukocyte infiltration, with neutrophils and macrophages responsible for eliminating microbes via phagocytosis [70]. If the invading pathogen was previously encountered, then cells of the adaptive immune response (i.e., T and B cells) also contribute to pathogen clearance [70].

Unfortunately, many pathogens possess tools that allow them to disrupt immune cell signaling processes. A common strategy among different bacteria is to interrupt phagocytosis, either by evading cellular uptake or by inhibiting phagolysosome activity [70-72]. Examples of bacteria that can disrupt phagocytosis include clinically significant pathogens, such as Mycobacterium tuberculosis, Streptococcus pyogenes, and Neisseria gonorrhoeae [72]. Because phagocytosis is the primary mechanism by which bacteria are eliminated from tissues, therapies that improve cellular phagocytic pathways can improve bacterial clearance [73,74]. In vitro studies of rodent derived macrophages have shown that ATRA increases phagocytic activity [27,75,76].

When applied to human HL60 cells (i.e., promyelocyte cell-line), both ATRA and dimethyl sulfoxide induce cellular differentiation into neutrophils [77]. However, only ATRA differentiated neutrophils exhibit increased phagocytic activity against bacteria, such as N. gonorrhoeae, Escherichia coli, and S. pyogenes [78,79]. Increased phagocytosis was associated with upregulated expression of carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), or CD66a [78]. CEACAM1 is a multifaceted membrane receptor that facilitates extracellular signal transduction to regulate different cellular functions (e.g., motility, proliferation, apoptosis, etc.) [80]. Important for immune function, CEACAM1 promotes monocyte survival and neutrophil function [80,81]. In neutrophils, CEACAM1 acts as a gram-negative bacterial receptor and promotes phagocytosis, especially against N. gonorrhoeae [80,82-84].

Along with facilitating leukocyte activity, ATRA can enhance antimicrobial defense in skin tissue. As the body’s primary defense against infection, skin tissue protects against pathogen invasion by acting as a barrier and by producing antimicrobial proteins (AMPs) [85]. One notable AMP is cathelicidin, which is a bactericidal compound that disrupts bacterial membranes and facilitates leukocyte recruitment [85-87]. Cathelicidin is encoded by the human CAMP gene (i.e., CRAMP gene in mice) and is secreted by keratinocytes, sebocytes, adipocytes, neutrophils, mast cells, and dendritic cells [86]. In mouse Staphylococcus aureus infection models, cathelicidin secretion by dermal adipocytes was shown to be crucial for limiting bacterial invasion [85,86,88]. When administered ATRA, preadipocytes exhibited increased cathelicidin secretion that reduced S. aureus growth [89].

ATRA’s antibacterial properties against M. tuberculosis (i.e., causative agent of tuberculosis) have been extensively studied and include different mechanisms of action [27,28]. HDTs against M. tuberculosis are especially important because of its increasing rate of antibiotic resistance [30,90]. Unlike other bacterial infections, Tuberculosis requires a monthslong multi-drug regimen (e.g., rifampicin, isoniazid, pyrazinamide, and ethambutol) for successful treatment [90]. With growing resistance to front-line antibiotics, ATRA’s antimicrobial effect against M. tuberculosis has received the most attention. In macrophages, ATRA improves M. tuberculosis clearance by promoting autophagy of engulfed bacteria [27,91,92]. Improved autophagy is crucial for M. tuberculosis clearance since it can survive inside macrophages by disrupting phagolysosome formation [72]. ATRA’s effect on macrophage autophagy stems from its activation of TANK-binding kinase 1 (TBK1), which localizes engulfed M. tuberculosis with autophagosomes [92,93]. Although TBK1 regulates the antiviral response, M. tuberculosis infection stimulates intracellular DNA PRRs that initiate an inflammatory immune response [93,94] (Figure 4). In monocytes, ATRA induces clearance by downregulating total cholesterol and increasing lysosomal acidification, which occurs in response to upregulation of NPC2 gene expression [95].

Figure 4. ATRA’s antibacterial properties. ATRA upregulates cellular antibacterial pathways through various mechanisms: improving phagocytosis, facilitating autophagy, and promoting Cathilicidin secretion. Created with BioRender.com.

The efficacy of ATRA as an HDT for tuberculosis is currently under investigation. In vivo studies of tuberculosis in rats have shown that ATRA treatment reduces disease severity and decreases M. tuberculosis growth [96]. Clinical studies examining vitamin A supplementation in tuberculosis patients have shown no benefit during early disease course [97]. However, treatments using active metabolites of vitamin A, such as ATRA, have not yet been tested during tuberculosis infection.

ATRA upregulation of antiviral response

The type-I Interferon (IFN) response is the innate immune system’s primary antiviral defense. Recognition of viral antigens by intracellular PRRs leads to IFN regulatory factor 3 (IRF3) activation [94,98]. IRF3 then induces IFNβ secretion, which stimulates antiviral gene expression in an autocrine and paracrine manner [94,98]. IFNβ promotes downstream activation of IRF9, which induces expression of over 300 antiviral proteins, such as IFN-induced tetratricopeptide repeat protein (IFIT), Myxovirus resistance protein 1 (MxA), and IFN-inducible transmembrane protein (IFITM) [94,99-101]. These proteins provide a potent antiviral defense, capable of interfering with each stage of the viral replication cycle (i.e., entry, translation, replication, and egress) [100,101].

Like other pathogens, viruses have developed effective strategies for disrupting immune signaling pathways. A prevalent technique during viral infection is to interrupt activation of the type-I IFN response [102]. Viral proteins can shield the viral genome from PRR detection or inhibit downstream signaling proteins. For instance, measles virus delays the type-I IFN response by inactivating intracellular PRRs, such as RA inducible gene I (RIG-I) and Melanoma differentiation associated gene 5 (MDA5) [102,103]. Other clinically important viruses, such as SARS coronavirus 2 (SARS-CoV-2) and influenza A virus, disrupt downstream signaling by inhibiting IRF3 activation or IFNβ receptor activity [102,104-106].

Whereas clinicians have a large array of antibiotics to treat bacterial infections, their repertoire of antivirals is much smaller in comparison. To complement the antiviral treatment regimen, different HDTs that can promote the type-I IFN response have been investigated [28]. Chief amongst them is ATRA, which can induce protection against different viral pathogens in vitro (e.g., measles, mumps, hepatitis C virus, etc.) [3,107-109]. Although not clinically used as an antiviral therapeutic, vitamin A does improve survival rates in pediatric patients (<2yr old) diagnosed with measles [110].

ATRA’s antiviral effect stems from its capacity to upregulate upstream mediators of the type-I IFN response. ATRA treatment increases expression of RIG-I, IFNβ, IFNβ receptor, IRF1, and antiviral proteins [107-109] (Figure 5). As a PRR against viral RNA, increased RIG-I expression can improve detection against RNA viruses, such as measles or SARS-CoV-2 [107,111,112]. IRF1 is not classically associated with the antiviral response; however, it can facilitate activation of IRF3 [113,114]. As the main driver of IFNβ secretion, IRF3 activation is crucial for propagating the type-I IFN response. Recent studies have also shown that ATRA exhibits direct antiviral activity, capable of inhibiting reverse transcriptase and 3C-like protease in human immunodeficiency virus and SARS-CoV-2, respectively [115,116]. Further research of ATRA’s antiviral properties is required to validate its efficacy in in vivo systems. Likewise, studies illustrating ATRA’s limited effectiveness against H9N2 influenza virus highlight the need to determine which specific viral pathogens are susceptible to ATRA treatment [117].

Figure 5. ATRA’s antiviral effect. ATRA confers protection against viral infections by upregulating the type-I IFN response. Specifically, ATRA increases IFNβ, IFNβ receptor, and antiviral protein expression. Created with BioRender.com.

Challenges of ATRA as a disease treatment

Unlike other novel therapeutics, RA and its derivatives have a long history of clinical use against various diseases (e.g., acne vulgaris, psoriasis, APL, etc.) [118]. RA’s effectiveness stems from its capacity to correct dysregulated cellular signaling pathways. However, its activity in multiple signaling pathways carries a significant risk of side effects, especially in pregnant women [1,119,120]. Common side effects of RA treatment include skin irritation and dryness [119,120]. The skin manifestations are due to RA’s effect on cellular differentiation, with RA inducing early maturation of skin cells [119,120]. Acute liver injury is an uncommon side effect of RA treatment, which usually presents with a transient elevation in liver enzymes [1].

The most significant side effect of RA treatment is fetal malformation during pregnancy [1,119,120]. RA’s characterization as a teratogen stem from its regulatory role during fetal development [1,4]. The addition of RA treatment during pregnancy alters normal fetal limb development and results in numerous malformations. Because it’s a teratogen, RA is never given during pregnancy. In women who require long term treatment with RA, such as in psoriasis, contraceptives are prescribed to reduce the risk of pregnancy. Also, topical RA therapeutics may be used because they act locally and are less likely to be absorbed systemically [120].

Conclusions

RA is a dynamic compound that is capable of inducing various cell signaling pathways. In APL, ATRA functions as an effective anti-cancer therapy by inducing cellular differentiation [36,40]. This process reduces cancer cell proliferation and promotes normal maturation. Its capacity to induce physiologic differentiation has been studied in different cancer cell-lines, with clinical benefits being seen against cutaneous T cell Lymphoma [44]. RA also exhibits an anti-inflammatory effect, which is used to treat psoriasis [56]. The ability of RA to downregulate inflammatory pathways, such as NF-κB, has highlighted its potential as a possible sepsis treatment [63]. Although clinical studies are needed to validate RA’s efficacy as a sepsis treatment, recent animal studies have illustrated its ability to improve sepsis survival rates [26].

The growing risk of antibiotic resistant disease and novel viral infection necessitates new therapeutic approaches. With decreasing numbers of approved antimicrobial treatments, HDTs have gained increasing importance as an alternative avenue for pharmaceutical research. Chief among them is ATRA, which can induce host antimicrobial pathways. Against bacterial pathogens, ATRA promotes phagocytosis, autophagy, cell survival, and AMP secretion (i.e., cathelicidin) [75,89,92]. In vitro studies with ATRA demonstrate that it improves clearance of clinically important pathogens, such as N. gonorrhoeae, S. aureus, and M. tuberculosis [78,89,91]. This effect is also seen against fungal pathogens, with ATRA stimulated macrophages exhibiting increased phagocytosis against Aspergillus fumigatus and Pneumocystis [121-123]. Against viral pathogens, ATRA upregulates the host type-I IFN response to promote viral clearance (e.g., measles, mumps, and hepatitis C virus) [107-109]. Taken together, RA’s capacity to alter cell signaling in different diseases has shown positive benefits that may be applicable to other pathologies.

Acknowledgements

We would like to thank our colleagues in the department of Medical Microbiology and Immunology for their advice and steadfast support.

References

1. Vitamins. LiverTox: Clinical and Research Information on Drug-Induced Liver Injury. Bethesda (MD), 2012.

2. Bollag W. Vitamin A and retinoids: from nutrition to pharmacotherapy in dermatology and oncology. Lancet. 1983;1(8329):860-3.

3. Pino-Lagos K, Guo Y, Noelle RJ. Retinoic acid: a key player in immunity. Biofactors. 2010;36(6):430-6.

4. Oliveira E, Casado M, Raldua D, Soares A, Barata C, Pina B. Retinoic acid receptors' expression and function during zebrafish early development. J Steroid Biochem Mol Biol. 2013;138:143-51.

5. Saari JC. Vitamin A and Vision. Subcell Biochem. 2016;81:231-59.

6. Hodge C, Taylor C. Vitamin A Deficiency. Treasure Island (FL): StatPearls; 2023.

7. Carazo A, Macakova K, Matousova K, Krcmova LK, Protti M, Mladenka P. Vitamin A Update: Forms, Sources, Kinetics, Detection, Function, Deficiency, Therapeutic Use and Toxicity. Nutrients. 2021 May 18;13(5):1703.

8. Harrison EH. Carotenoids, beta-Apocarotenoids, and Retinoids: The Long and the Short of It. Nutrients. 2022;14(7):1411.

9. O'Byrne SM, Blaner WS. Retinol and retinyl esters: biochemistry and physiology. J Lipid Res. 2013;54(7):1731-43.

10. Napoli JL. Retinoic acid biosynthesis and metabolism. FASEB J. 1996;10(9):993-1001.

11. Njar VC, Gediya L, Purushottamachar P, Chopra P, Vasaitis TS, Khandelwal A, et al. Retinoic acid metabolism blocking agents (RAMBAs) for treatment of cancer and dermatological diseases. Bioorg Med Chem. 2006;14(13):4323-40.

12. le Maire A, Teyssier C, Balaguer P, Bourguet W, Germain P. Regulation of RXR-RAR Heterodimers by RXR- and RAR-Specific Ligands and Their Combinations. Cells. 2019;8(11):1392.

13. Kersten S, Gronemeyer H, Noy N. The DNA binding pattern of the retinoid X receptor is regulated by ligand-dependent modulation of its oligomeric state. J Biol Chem. 1997;272(19):12771-7.

14. Al Tanoury Z, Piskunov A, Rochette-Egly C. Vitamin A and retinoid signaling: genomic and nongenomic effects. J Lipid Res. 2013;54(7):1761-75.

15. Balmer JE, Blomhoff R. Gene expression regulation by retinoic acid. J Lipid Res. 2002;43(11):1773-808.

16. Penvose A, Keenan JL, Bray D, Ramlall V, Siggers T. Comprehensive study of nuclear receptor DNA binding provides a revised framework for understanding receptor specificity. Nat Commun. 2019;10(1):2514.

17. Wang ZY, Chen Z. Acute promyelocytic leukemia: from highly fatal to highly curable. Blood. 2008;111(5):2505-15.

18. Liang C, Qiao G, Liu Y, Tian L, Hui N, Li J, et al. Overview of all-trans-retinoic acid (ATRA) and its analogues: Structures, activities, and mechanisms in acute promyelocytic leukaemia. Eur J Med Chem. 2021;220:113451.

19. Altucci L, Gronemeyer H. The promise of retinoids to fight against cancer. Nat Rev Cancer. 2001;1(3):181-93.

20. World Health Organization Model List of Essential Medicines: World Health Organization; 2021.

21. Moreb JS, Ucar-Bilyeu DA, Khan A. Use of retinoic acid/aldehyde dehydrogenase pathway as potential targeted therapy against cancer stem cells. Cancer Chemother Pharmacol. 2017;79(2):295-301.

22. Chen MC, Hsu SL, Lin H, Yang TY. Retinoic acid and cancer treatment. Biomedicine (Taipei). 2014;4(4):22.

23. Szymanski L, Skopek R, Palusinska M, Schenk T, Stengel S, Lewicki S, et al. Retinoic Acid and Its Derivatives in Skin. Cells. 2020;9(12):2660.

24. Austenaa LM, Carlsen H, Hollung K, Blomhoff HK, Blomhoff R. Retinoic acid dampens LPS-induced NF-kappaB activity: results from human monoblasts and in vivo imaging of NF-kappaB reporter mice. J Nutr Biochem. 2009;20(9):726-34.

25. Kim BH, Kang KS, Lee YS. Effect of retinoids on LPS-induced COX-2 expression and COX-2 associated PGE(2) release from mouse peritoneal macrophages and TNF-alpha release from rat peripheral blood mononuclear cells. Toxicol Lett. 2004;150(2):191-201.

26. Dolin HH, Franco JH, Chen X, Pan ZK. Retinoic Acid-Induced Regulation of Inflammatory Pathways Is a Potential Sepsis Treatment. Infect Immun. 2023;91(4):e0045722.

27. Bahlool AZ, Grant C, Cryan SA, Keane J, O'Sullivan MP. All trans retinoic acid as a host-directed immunotherapy for tuberculosis. Curr Res Immunol. 2022;3:54-72.

28. Kaufmann SHE, Dorhoi A, Hotchkiss RS, Bartenschlager R. Host-directed therapies for bacterial and viral infections. Nat Rev Drug Discov. 2018;17(1):35-56.

29. Davies J, Davies D. Origins and evolution of antibiotic resistance. Microbiol Mol Biol Rev. 2010;74(3):417-33.

30. Antimicrobial Resistance Collaborators. Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. Lancet. 2022;399(10325):629-55.

31. Brandenburg K, Schurholz T. Lack of new antiinfective agents: Passing into the pre-antibiotic age? World J Biol Chem. 2015;6(3):71-7.

32. Mercorelli B, Palu G, Loregian A. Drug Repurposing for Viral Infectious Diseases: How Far Are We? Trends Microbiol. 2018;26(10):865-76.

33. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646-74.

34. Kontomanolis EN, Koutras A, Syllaios A, Schizas D, Mastoraki A, Garmpis N, et al. Role of Oncogenes and Tumor-suppressor Genes in Carcinogenesis: A Review. Anticancer Res. 2020;40(11):6009-15.

35. Joyce C, Rayi A, Kasi A. Tumor-Suppressor Genes. Treasure Island (FL): StatPearls; 2023.

36. Yilmaz M, Kantarjian H, Ravandi F. Acute promyelocytic leukemia current treatment algorithms. Blood Cancer J. 2021;11(6):123.

37. Jimenez JJ, Chale RS, Abad AC, Schally AV. Acute promyelocytic leukemia (APL): a review of the literature. Oncotarget. 2020;11(11):992-1003.

38. Dzierzak E, Philipsen S. Erythropoiesis: development and differentiation. Cold Spring Harb Perspect Med. 2013;3(4):a011601.

39. Cheng H, Zheng Z, Cheng T. New paradigms on hematopoietic stem cell differentiation. Protein Cell. 2020;11(1):34-44.

40. Warrell RP, Jr., Frankel SR, Miller WH, Jr., Scheinberg DA, Itri LM, Hittelman WN, et al. Differentiation therapy of acute promyelocytic leukemia with tretinoin (all-trans-retinoic acid). N Engl J Med. 1991;324(20):1385-93.

41. Zhou W, Bao S. PML-mediated signaling and its role in cancer stem cells. Oncogene. 2014;33(12):1475-84.

42. Guan D, Kao HY. The function, regulation and therapeutic implications of the tumor suppressor protein, PML. Cell Biosci. 2015;5:60.

43. Nowak D, Stewart D, Koeffler HP. Differentiation therapy of leukemia: 3 decades of development. Blood. 2009;113(16):3655-65.

44. Schadt CR. Topical and oral bexarotene. Dermatol Ther. 2013;26(5):400-3.

45. Hunsu VO, Facey COB, Fields JZ, Boman BM. Retinoids as Chemo-Preventive and Molecular-Targeted Anti-Cancer Therapies. Int J Mol Sci. 2021;22(14):7731.

46. Sporn MB, Suh N. Chemoprevention of cancer. Carcinogenesis. 2000;21(3):525-30.

47. Femiano F, Gombos F, Scully C, Battista C, Belnome G, Esposito V. Oral leukoplakia: open trial of topical therapy with calcipotriol compared with tretinoin. Int J Oral Maxillofac Surg. 2001;30(5):402-6.

48. Kraemer KH, DiGiovanna JJ, Moshell AN, Tarone RE, Peck GL. Prevention of skin cancer in xeroderma pigmentosum with the use of oral isotretinoin. N Engl J Med. 1988;318(25):1633-7.

49. Mrass P, Rendl M, Mildner M, Gruber F, Lengauer B, Ballaun C, et al. Retinoic acid increases the expression of p53 and proapoptotic caspases and sensitizes keratinocytes to apoptosis: a possible explanation for tumor preventive action of retinoids. Cancer Res. 2004;64(18):6542-8.

50. Chen XJ, He MJ, Zhou G. All-trans retinoic acid induces anti-tumor effects via STAT3 signaling inhibition in oral squamous cell carcinoma and oral dysplasia. J Oral Pathol Med. 2019;48(9):832-9.

51. Chen L, Deng H, Cui H, Fang J, Zuo Z, Deng J, et al. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget. 2018;9(6):7204-18.

52. Akdis M, Aab A, Altunbulakli C, Azkur K, Costa RA, Crameri R, et al. Interleukins (from IL-1 to IL-38), interferons, transforming growth factor beta, and TNF-alpha: Receptors, functions, and roles in diseases. J Allergy Clin Immunol. 2016;138(4):984-1010.

53. Liu T, Zhang L, Joo D, Sun SC. NF-kappaB signaling in inflammation. Signal Transduct Target Ther. 2017;2:17023.

54. Kim EK, Choi EJ. Pathological roles of MAPK signaling pathways in human diseases. Biochim Biophys Acta. 2010;1802(4):396-405.

55. Griffiths CE, Barker JN. Pathogenesis and clinical features of psoriasis. Lancet. 2007;370(9583):263-71.

56. Tokuyama M, Mabuchi T. New Treatment Addressing the Pathogenesis of Psoriasis. Int J Mol Sci. 2020;21(20):7488.

57. Ogawa E, Sato Y, Minagawa A, Okuyama R. Pathogenesis of psoriasis and development of treatment. J Dermatol. 2018;45(3):264-72.

58. Rafieian-Kopaei M, Setorki M, Doudi M, Baradaran A, Nasri H. Atherosclerosis: process, indicators, risk factors and new hopes. Int J Prev Med. 2014;5(8):927-46.

59. Deng Q, Chen J. Potential Therapeutic Effect of All-Trans Retinoic Acid on Atherosclerosis. Biomolecules. 2022;12(7):869.

60. Mehta K, McQueen T, Tucker S, Pandita R, Aggarwal BB. Inhibition by all-trans-retinoic acid of tumor necrosis factor and nitric oxide production by peritoneal macrophages. J Leukoc Biol. 1994;55(3):336-42.

61. Na SY, Kang BY, Chung SW, Han SJ, Ma X, Trinchieri G, et al. Retinoids inhibit interleukin-12 production in macrophages through physical associations of retinoid X receptor and NFkappaB. J Biol Chem. 1999;274(12):7674-80.

62. Benson MJ, Pino-Lagos K, Rosemblatt M, Noelle RJ. All-trans retinoic acid mediates enhanced T reg cell growth, differentiation, and gut homing in the face of high levels of co-stimulation. J Exp Med. 2007;204(8):1765-74.

63. Franco JH, Chen X, Pan ZK. Novel Treatments Targeting the Dysregulated Cell Signaling Pathway during Sepsis. J Cell Signal. 2021;2(4):228-34.

64. Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016;315(8):801-10.

65. Font MD, Thyagarajan B, Khanna AK. Sepsis and Septic Shock - Basics of diagnosis, pathophysiology and clinical decision making. Med Clin North Am. 2020;104(4):573-85.

66. Hotchkiss RS, Moldawer LL, Opal SM, Reinhart K, Turnbull IR, Vincent JL. Sepsis and septic shock. Nat Rev Dis Primers. 2016;2:16045.

67. Dolin HH, Papadimos TJ, Chen X, Pan ZK. Characterization of Pathogenic Sepsis Etiologies and Patient Profiles: A Novel Approach to Triage and Treatment. Microbiol Insights. 2019;12:1178636118825081.

68. Jain S. Sepsis: An Update on Current Practices in Diagnosis and Management. Am J Med Sci. 2018;356(3):277-86.

69. Deutschman CS, Tracey KJ. Sepsis: current dogma and new perspectives. Immunity. 2014;40(4):463-75.

70. Justiz Vaillant AA, Sabir S, Jan A. Physiology, Immune Response. Treasure Island (FL): StatPearls; 2023.

71. Leseigneur C, Le-Bury P, Pizarro-Cerda J, Dussurget O. Emerging Evasion Mechanisms of Macrophage Defenses by Pathogenic Bacteria. Front Cell Infect Microbiol. 2020;10:577559.

72. Uribe-Querol E, Rosales C. Control of Phagocytosis by Microbial Pathogens. Front Immunol. 2017;8:1368.

73. Mandell GL. Cytokines, phagocytes, and pentoxifylline. J Cardiovasc Pharmacol. 1995;25 Suppl 2:S20-2.

74. Gierlikowska B, Stachura A, Gierlikowski W, Demkow U. Phagocytosis, Degranulation and Extracellular Traps Release by Neutrophils-The Current Knowledge, Pharmacological Modulation and Future Prospects. Front Pharmacol. 2021;12:666732.

75. Goldman R. Effect of retinoic acid on the proliferation and phagocytic capability of murine macrophage-like cell lines. J Cell Physiol. 1984;120(1):91-102.

76. Li S, Lei Y, Lei J, Li H. All-trans retinoic acid promotes macrophage phagocytosis and decreases inflammation via inhibiting CD14/TLR4 in acute lung injury. Mol Med Rep. 2021;24(6):868.

77. Birnie GD. The HL60 cell line: a model system for studying human myeloid cell differentiation. Br J Cancer Suppl. 1988;9:41-5.

78. Pantelic M, Chen I, Parker J, Zhang P, Grunert F, Chen T. Retinoic acid treated HL60 cells express CEACAM1 (CD66a) and phagocytose Neisseria gonorrhoeae. FEMS Immunol Med Microbiol. 2004;42(2):261-6.

79. Nordenfelt P, Bauer S, Lonnbro P, Tapper H. Phagocytosis of Streptococcus pyogenes by all-trans retinoic acid-differentiated HL-60 cells: roles of azurophilic granules and NADPH oxidase. PLoS One. 2009;4(10):e7363.

80. Kim WM, Huang YH, Gandhi A, Blumberg RS. CEACAM1 structure and function in immunity and its therapeutic implications. Semin Immunol. 2019;42:101296.

81. Yu Q, Chow EM, Wong H, Gu J, Mandelboim O, Gray-Owen SD, et al. CEACAM1 (CD66a) promotes human monocyte survival via a phosphatidylinositol 3-kinase- and AKT-dependent pathway. J Biol Chem. 2006;281(51):39179-93.

82. Voges M, Bachmann V, Kammerer R, Gophna U, Hauck CR. CEACAM1 recognition by bacterial pathogens is species-specific. BMC Microbiol. 2010;10:117.

83. Gray-Owen SD, Dehio C, Haude A, Grunert F, Meyer TF. CD66 carcinoembryonic antigens mediate interactions between Opa-expressing Neisseria gonorrhoeae and human polymorphonuclear phagocytes. EMBO J. 1997;16(12):3435-45.

84. Sarantis H, Gray-Owen SD. Defining the roles of human carcinoembryonic antigen-related cellular adhesion molecules during neutrophil responses to Neisseria gonorrhoeae. Infect Immun. 2012;80(1):345-58.

85. Kwiecien K, Zegar A, Jung J, Brzoza P, Kwitniewski M, Godlewska U, et al. Architecture of antimicrobial skin defense. Cytokine Growth Factor Rev. 2019;49:70-84.

86. Zhang LJ, Gallo RL. Antimicrobial peptides. Curr Biol. 2016;26(1):R14-9.

87. Alford MA, Baquir B, Santana FL, Haney EF, Hancock REW. Cathelicidin Host Defense Peptides and Inflammatory Signaling: Striking a Balance. Front Microbiol. 2020;11:1902.

88. Zhang LJ, Guerrero-Juarez CF, Hata T, Bapat SP, Ramos R, Plikus MV, et al. Innate immunity. Dermal adipocytes protect against invasive Staphylococcus aureus skin infection. Science. 2015;347(6217):67-71.

89. Liggins MC, Li F, Zhang LJ, Dokoshi T, Gallo RL. Retinoids Enhance the Expression of Cathelicidin Antimicrobial Peptide during Reactive Dermal Adipogenesis. J Immunol. 2019;203(6):1589-97.

90. Singh R, Dwivedi SP, Gaharwar US, Meena R, Rajamani P, Prasad T. Recent updates on drug resistance in Mycobacterium tuberculosis. J Appl Microbiol. 2020;128(6):1547-67.

91. Crowle AJ, Ross EJ. Inhibition by retinoic acid of multiplication of virulent tubercle bacilli in cultured human macrophages. Infect Immun. 1989;57(3):840-4.

92. Coleman MM, Basdeo SA, Coleman AM, Cheallaigh CN, Peral de Castro C, McLaughlin AM, et al. All-trans Retinoic Acid Augments Autophagy during Intracellular Bacterial Infection. Am J Respir Cell Mol Biol. 2018;59(5):548-56.

93. Watson RO, Manzanillo PS, Cox JS. Extracellular M. tuberculosis DNA targets bacteria for autophagy by activating the host DNA-sensing pathway. Cell. 2012;150(4):803-15.

94. Franco JH, Chattopadhyay S, Pan ZK. How Different Pathologies Are Affected by IFIT Expression. Viruses. 2023;15(2):342.

95. Wheelwright M, Kim EW, Inkeles MS, De Leon A, Pellegrini M, Krutzik SR, et al. All-trans retinoic acid-triggered antimicrobial activity against Mycobacterium tuberculosis is dependent on NPC2. J Immunol. 2014;192(5):2280-90.

96. Yamada H, Mizuno S, Ross AC, Sugawara I. Retinoic acid therapy attenuates the severity of tuberculosis while altering lymphocyte and macrophage numbers and cytokine expression in rats infected with Mycobacterium tuberculosis. J Nutr. 2007;137(12):2696-700.

97. Visser ME, Grewal HM, Swart EC, Dhansay MA, Walzl G, Swanevelder S, et al. The effect of vitamin A and zinc supplementation on treatment outcomes in pulmonary tuberculosis: a randomized controlled trial. Am J Clin Nutr. 2011;93(1):93-100.

98. Negishi H, Taniguchi T, Yanai H. The Interferon (IFN) Class of Cytokines and the IFN Regulatory Factor (IRF) Transcription Factor Family. Cold Spring Harb Perspect Biol. 2018;10(11):a028423.

99. Schoggins JW. Interferon-Stimulated Genes: What Do They All Do? Annu Rev Virol. 2019;6(1):567-84.

100. Schoggins JW. Interferon-stimulated genes: roles in viral pathogenesis. Curr Opin Virol. 2014;6:40-6.

101. Schneider WM, Chevillotte MD, Rice CM. Interferon-stimulated genes: a complex web of host defenses. Annu Rev Immunol. 2014;32:513-45.

102. Garcia-Sastre A. Ten Strategies of Interferon Evasion by Viruses. Cell Host Microbe. 2017;22(2):176-84.

103. Davis ME, Wang MK, Rennick LJ, Full F, Gableske S, Mesman AW, et al. Antagonism of the phosphatase PP1 by the measles virus V protein is required for innate immune escape of MDA5. Cell Host Microbe. 2014;16(1):19-30.

104. Zhang Q, Chen Z, Huang C, Sun J, Xue M, Feng T, et al. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Membrane (M) and Spike (S) Proteins Antagonize Host Type I Interferon Response. Front Cell Infect Microbiol. 2021;11:766922.

105. Du Y, Yang F, Wang Q, Xu N, Xie Y, Chen S, et al. Influenza a virus antagonizes type I and type II interferon responses via SOCS1-dependent ubiquitination and degradation of JAK1. Virol J. 2020;17(1):74.

106. Borrow P, Martinez-Sobrido L, de la Torre JC. Inhibition of the type I interferon antiviral response during arenavirus infection. Viruses. 2010;2(11):2443-80.

107. Soye KJ, Trottier C, Richardson CD, Ward BJ, Miller WH, Jr. RIG-I is required for the inhibition of measles virus by retinoids. PLoS One. 2011;6(7):e22323.

108. Soye KJ, Trottier C, Di Lenardo TZ, Restori KH, Reichman L, Miller WH, Jr., et al. In vitro inhibition of mumps virus by retinoids. Virol J. 2013;10:337.

109. Hamamoto S, Fukuda R, Ishimura N, Rumi MA, Kazumori H, Uchida Y, et al. 9-cis retinoic acid enhances the antiviral effect of interferon on hepatitis C virus replication through increased expression of type I interferon receptor. J Lab Clin Med. 2003;141(1):58-66.

110. D'Souza RM, D'Souza R. Vitamin A for treating measles in children. Cochrane Database Syst Rev. 2002(1):CD001479.

111. Li D, Wu M. Pattern recognition receptors in health and diseases. Signal Transduct Target Ther. 2021;6(1):291.

112. Kouwaki T, Nishimura T, Wang G, Oshiumi H. RIG-I-Like Receptor-Mediated Recognition of Viral Genomic RNA of Severe Acute Respiratory Syndrome Coronavirus-2 and Viral Escape From the Host Innate Immune Responses. Front Immunol. 2021;12:700926.

113. Antonczyk A, Krist B, Sajek M, Michalska A, Piaszyk-Borychowska A, Plens-Galaska M, et al. Direct Inhibition of IRF-Dependent Transcriptional Regulatory Mechanisms Associated With Disease. Front Immunol. 2019;10:1176.

114. Wang J, Li H, Xue B, Deng R, Huang X, Xu Y, et al. IRF1 Promotes the Innate Immune Response to Viral Infection by Enhancing the Activation of IRF3. J Virol. 2020;94(22) ):e01231-20.

115. Maeda Y, Yamaguchi T, Hijikata Y, Morita Y, Tanaka M, Hirase C, et al. All-trans retinoic acid attacks reverse transcriptase resulting in inhibition of HIV-1 replication. Hematology. 2007;12(3):263-6.

116. Morita T, Miyakawa K, Jeremiah SS, Yamaoka Y, Sada M, Kuniyoshi T, et al. All-Trans Retinoic Acid Exhibits Antiviral Effect against SARS-CoV-2 by Inhibiting 3CLpro Activity. Viruses. 2021;13(8):1669.

117. Niu X, Wang H, Zhao L, Lian P, Bai Y, Li J, et al. All-trans retinoic acid increases the pathogenicity of the H9N2 influenza virus in mice. Virol J. 2022;19(1):113.

118. Baldwin HE, Nighland M, Kendall C, Mays DA, Grossman R, Newburger J. 40 years of topical tretinoin use in review. J Drugs Dermatol. 2013;12(6):638-42.

119. Yob EH, Pochi PE. Side effects and long-term toxicity of synthetic retinoids. Arch Dermatol. 1987;123(10):1375-8.

120. Motamedi M, Chehade A, Sanghera R, Grewal P. A Clinician's Guide to Topical Retinoids. J Cutan Med Surg. 2022;26(1):71-8.

121. Cosio T, Gaziano R, Zuccari G, Costanza G, Grelli S, Di Francesco P, et al. Retinoids in Fungal Infections: From Bench to Bedside. Pharmaceuticals (Basel). 2021;14(10):962.

122. Campione E, Gaziano R, Doldo E, Marino D, Falconi M, Iacovelli F, et al. Antifungal Effect of All-trans Retinoic Acid against Aspergillus fumigatus In Vitro and in a Pulmonary Aspergillosis In Vivo Model. Antimicrob Agents Chemother. 2021;65(3):e01874-20.

123. Lei GS, Zhang C, Shao S, Jung HW, Durant PJ, Lee CH. All-trans retinoic acid in combination with primaquine clears pneumocystis infection. PLoS One. 2013;8(1):e53479.

Author Information X