Skip to content
BY 4.0 license Open Access Published by De Gruyter December 30, 2021

SIT1 transporter as a potential novel target in treatment of COVID-19

  • Sabina Semiz EMAIL logo
From the journal Biomolecular Concepts

Abstract

Studies published earlier this year demonstrated the association of the solute carrier SLC6A20 gene with the risk and severity of COVID-19. The SLC6A20 protein product (Sodium-dependent Imino Transporter 1 (SIT1)) is involved in the transport of amino acids, including glycine. Here we summarized the results of recent studies demonstrating the interaction of SIT1 with the ACE2 receptor for SARS-CoV-2 as well as an observed association of SLC6A20 with the risk and traits of Type 2 diabetes (T2D). Recently, it was also proposed that SLC6A20 represents the novel regulator of glycine levels and that glycine has beneficial effects against the proinflammatory cytokine secretion induced by SARS-CoV-2 infection. Ivermectin, as a partial agonist of glycine-gated chloride channels, was also recently suggested to interfere with the COVID-19 cytokine storm by inducing the activation of glycine receptors. Furthermore, plasma glycine levels are found to be decreased in diabetic patients. Thus, further clinical trials are warranted to confirm the potential favorable effects of targeting the SIT1 transporter and glycine levels in the treatment of COVID-19, particularly for the severe case of disease associated with hyperglycemia, inflammation, and T2D. These findings suggest that SIT1 may potentially represent one of the missing pieces in the complex puzzle observed between these two pandemic diseases and the potential novel target for their efficient treatment.

Introduction

In search of potential genetic factors associated with the development of the coronavirus disease 2019 (COVID-19), studies published earlier this year identified the solute carrier SLC6A20 as one of the few genes associated with the risk and severity of COVID-19 [1,2]. The protein product of SLC6A20 is Sodium-dependent Imino Transporter 1 (System IMINO transporter (SIT1)), which is involved in the transport of amino acids, including glycine [3] and proline [4]. Interestingly, the SIT1 transporter is reported to co-express with the angiotensin-converting enzyme 2 (ACE2) receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the human small intestine [5,6]. It is also demonstrated that SIT1 heterodimerizes with ACE2, which appears to be required for formation of quaternary structures able to function as binding sites for SARS-CoV-2 spike glycoproteins [7]. Thus, these findings indicate that the interaction of the SIT1 transporter with the ACE2 receptor is required for viral entry and the consequent development of SARS-Cov-2 infection. The modes of SIT1 and ACE2 interaction, particularly in the case of severe COVID-19 associated with hyperglycemia and other traits of Type 2 diabetes (T2D), are still not well understood and the potential mechanisms are discussed in the chapters below.

Association of SIT1 and COVID-19

In addition to the recent reports of the association of the solute carrier SLC6A20 gene with the risk and severity of COVID-19 [1,2], the results of the genome-wide association study (GWAS) demonstrated that the SLC6A20 gene, located in an intronic region of chromosome 3, locus 3p21.31, with the minor rs11385942(A) allele, yielded the strongest association signal across the genome for COVID-19 respiratory failure [8] (Table 1). Furthermore, it was reported that SLC6A20 is predominantly expressed in alveolar type 2 cells and that changes in the SLC6A20 expression in these cells appear to impact the severity of the infection of the lungs with SARS-CoV-2 [9].

Table 1

Relevant publications dealing with association of SLC6A20/SIT1 with COVID-19 and/or ACE2 receptor for SARS-CoV-2.

SIT1 association with COVID-19 and ACE2 receptor Type of Study References
Risk and severity of COVID-19 GWAS (1, 2)
COVID-19 respiratory failure GWAS (8)
SIT1 co-localize with ACE2 receptor in human small intestine; ACE2 expression promotes SIT1 levels In vitro (human small intestine enterocytes) (6)
SIT1 co-express with ACE2 in intestine In vivo animal (ACE2-null mice) (10)

Previous studies indicated the functional interaction of SIT1 with key players involved in the SARS-CoV-2 infection, including its ACE2 receptor [6,10]. The SIT1 transporter and ACE2 receptor were reported to co-localize in the human small intestine [5,6]. The authors showed that ACE2 expression promotes SIT1 levels, its localization to plasma membrane and its function in amino acid transport [6]. Furthermore, it was demonstrated that ACE2 protein heterodimerizes with SLC6A20/SIT1 and its close relative SLC6A19, encoding the Broad neutral Amino acid Transporter 1 (B0AT1), which seems to enable these transporters to function as binding sites for SARS-CoV-2 spike glycoproteins [7]. Thus, these findings suggest that in addition to targeting the ACE2 receptor as recently suggested [11], SIT1 may also represent a potential novel target for COVID-19 treatment.

COVID-19 and Type 2 diabetes

The evidence has been accumulating to demonstrate that patients with severe COVID-19 are more likely to have a history of diabetes, hypertension, and/or cardiovascular disease [12,13,14]. The diabetic patients are exposed to an enhanced risk of serious complications, such as cardiac arrest [15,16], neurological disorders [17,18], and venous thromboembolism [19], as well as to an increased severity of this devastating disease [20]. A chronic low-grade inflammation and a more rapid ageing of the immune system have been suggested as the potential mechanisms associated with the higher susceptibility to serious COVID-19 outcomes in T2D patients [21,22]. Furthermore, it was shown that patients with a more severe progression of diabetes seem to have a worse prognosis of COVID-19 as compared to diabetic patients with a milder stage of disease [23]. In addition, it was reported that uncontrolled hyperglycemia augments the risk of poor prognosis [24] and may be a strong predictor of mortality and adverse outcomes in COVID-19 patients [25,26,27,28]. Figure 1 illustrates the potential components/mechanisms having a role in COVID-19 and Type 2 diabetes puzzling interconnection, whose roles and regulation are still not completely understood.

Figure 1 Schematic diagram of the potential common components/mechanisms involved in pathogenesis of both, coronavirus disease 2019 (COVID-19) and Type 2 diabetes mellitus (T2D). COVID-19 may predispose the patients to difficult-to-treat hyperglycemia (51, 85) as well as to insulin resistance and impaired insulin secretion (86), which are known T2D traits; A chronic low-grade inflammation, increased levels of inflammatory cytokines, and a perturbed immune system response are also associated with both of these diseases (21, 22, 87, 88); Furthermore, infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) stimulates the production of reactive oxygen species (ROS) (20, 89), which is also an established factor associated with T2D (90), vascular endothelial damage, thromboembolism, and cardiovascular complications often present in serious COVID-19 (20); The modulation of the renin–angiotensin–aldosterone system (RAAS) has been associated with hypertension, inflammation, thrombosis, and myocarditis in COVID-19 patients (91); It is established that RAAS inhibition is beneficial in preventing new onset of T2D (92); In addition, decreased glycine levels are also associated with T2D (79–81). Glycine has demonstrated beneficial effects against the inflammatory cytokine secretion induced by SARS-CoV-2 infection (53).
Figure 1

Schematic diagram of the potential common components/mechanisms involved in pathogenesis of both, coronavirus disease 2019 (COVID-19) and Type 2 diabetes mellitus (T2D). COVID-19 may predispose the patients to difficult-to-treat hyperglycemia (51, 85) as well as to insulin resistance and impaired insulin secretion (86), which are known T2D traits; A chronic low-grade inflammation, increased levels of inflammatory cytokines, and a perturbed immune system response are also associated with both of these diseases (21, 22, 87, 88); Furthermore, infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) stimulates the production of reactive oxygen species (ROS) (20, 89), which is also an established factor associated with T2D (90), vascular endothelial damage, thromboembolism, and cardiovascular complications often present in serious COVID-19 (20); The modulation of the renin–angiotensin–aldosterone system (RAAS) has been associated with hypertension, inflammation, thrombosis, and myocarditis in COVID-19 patients (91); It is established that RAAS inhibition is beneficial in preventing new onset of T2D (92); In addition, decreased glycine levels are also associated with T2D (79–81). Glycine has demonstrated beneficial effects against the inflammatory cytokine secretion induced by SARS-CoV-2 infection (53).

It was recommended that patients with diabetes and COVID-19 should be closely monitored and adequately treated to prevent or minimize the development of the potential deleterious effects associated with the co-presence of these diseases [29]. Interestingly, recent studies showed that metformin treatment of Type 2 diabetic patients was associated with reduced mortality for COVID-19 [30,31,32]. Different mechanisms are implicated so far among protective roles of metformin, including an improvement of insulin resistance [33], regulation of the blood glucose levels [34], regulation of the reninangiotensin-aldosterone system (RAAS) [35] and ACE2 receptor [34] that is a part of the RAAS [36], and decreasing levels of circulating cytokines [21,37].

Association of SIT1 and Type 2 Diabetes

Strikingly, Ling et al. demonstrated a highly significant association of the rs13062383 variant of the SLC6A20 gene with Type 2 diabetes in Caucasian and Chinese population [38] (Table 2). SLC6A20 has been reported to regulate the RAAS and water/salt reabsorption [38,39]. RAAS antagonists are known to reduce the risk of diabetes development [40,41] and improve glycemic control in diabetic patients [42], suggesting that the potential targeting of SLC6A20/SIT1 may perhaps produce beneficial effects in T2D patients by affecting the RAAS. Interestingly, the pharmacological targeting of SLC6A19/B0AT1 transporter, which is a close relative of SLC6A20/SIT1 transporter, has been recently suggested as a target in treatment of Type 2 diabetes [43]. Although decreased expression of SLC6A20 mRNA levels is reported in the kidneys of nonhuman primate and mouse models of T2D [44], further studies are pertinent to analyze the expression profile and regulation of SIT1 in diabetic patients.

Table 2

Relevant publications dealing with association of SLC6A20/SIT1 with Type 2 Diabetes (T2D).

SIT1 association with T2D risk and traits Type of Study References
SLC6A20 rs13062383 SNP associated with increased T2D risk Prospective, population-based study (38)
Decreased expression of SLC6A20 mRNA levels in T2D Nonhuman primate model of T2D (44)
SIT1 regulates glycine levels; (glycine levels are decreased in T2D (79–81)). In vivo animal (SLC6A20a−/− knockout mouse) (3)

As mentioned earlier, the SIT1 transporter and ACE2 receptor for SARS-CoV-2 were reported to co-localize and interact with each other in vitro [6]. Decreased kidney expression of ACE2 was also observed in patients with Type 2 diabetes [45,46], which was suggested to contribute to poor prognosis in COVID-19 patients [47]. Furthermore, ACE2 knockout mice were more prone to high-fat diet-induced pancreatic β-cell dysfunction and had impaired glucose tolerance/diabetes [48]. Since it was also shown that the activation of the ACE2/(A1-7)/Mas axis can improve insulin resistance and increase glucose uptake in the liver [49] as well as regulate the insulin/Akt signaling pathway, ACE2 was proposed to be a novel drug target for treating insulin resistance [50]. Furthermore, SIT1 (SLC6A20) and ACE2 may potentially regulate the mechanistic link associated with the severity of COVID-19 in Type 2 diabetic subjects [20] and difficult-to-treat hyperglycemia cases in COVID-19 patients [26,51].

Regulation of glycine levels in COVID-19

In order to further clarify the function of the SIT1 protein, earlier this year Bae et al. developed the SLC6A20a−/− knockout mouse model for SIT1 and reported that these knockout mice had a decreased survival rate [3]. Interestingly, the extracellular levels of brain glycine were strongly increased in the SLC6A20a−/− mice brain and the antisense knockdown of SLC6A20 expression also increased the glycine concentration in the brain [3], suggesting that SLC6A20 is a novel regulator of glycine levels which may play an important role in COVID-19 development (Figure 2).

Figure 2 Schematic representation of a potential role of SLC6A20 protein product SIT1 in regulation of glycine levels as well as in treatment of COVID-19 and Type 2 diabetes (T2D). The interaction of SIT1 with the ACE2 receptor for SARS-CoV-2 (6) as well as with the risk and traits of T2D (38) were proposed; The SIT1 transporter is involved in the transport of amino acids, including glycine (3), which appears to have beneficial effects against the proinflammatory cytokine secretion induced by SARS-CoV-2 infection (53) and in prognosis of T2D (82); Ivermectin, as a partial agonist of glycine-gated chloride channels, was recently suggested to interfere with the COVID-19 cytokine storm by inducing the activation of glycine receptors (GlyR) (77).
Figure 2

Schematic representation of a potential role of SLC6A20 protein product SIT1 in regulation of glycine levels as well as in treatment of COVID-19 and Type 2 diabetes (T2D). The interaction of SIT1 with the ACE2 receptor for SARS-CoV-2 (6) as well as with the risk and traits of T2D (38) were proposed; The SIT1 transporter is involved in the transport of amino acids, including glycine (3), which appears to have beneficial effects against the proinflammatory cytokine secretion induced by SARS-CoV-2 infection (53) and in prognosis of T2D (82); Ivermectin, as a partial agonist of glycine-gated chloride channels, was recently suggested to interfere with the COVID-19 cytokine storm by inducing the activation of glycine receptors (GlyR) (77).

Interestingly, the plasma levels of glycine appear to be significantly decreased in ACE2 null mice [52], suggesting that the activation of ACE2 would increase plasma glycine levels. Recently, it was proposed that glycine itself should be used in the treatment of COVID-19 patients due to its anti-inflammatory, anti-oxidative, neurological functions and role in metabolic regulation [53]. It was suggested that glycine, by binding to its receptor GlyR, induces a chloride influx causing cellular membrane hyperpolarization that guards the cells from pyroptosis and a proinflammatory cytokine storm resulting from SARS-CoV-2 infection [53].

One of the drugs that could be potentially repositioned for COVID-19 treatment and used to affect the glycine levels is ivermectin (IVM), which is known as a partial agonist of glycine-gated chloride channels [54,55]. Recently, IVM has become a subject of many controversies regarding its potential use for the treatment of COVID-19. Earlier this year it has been proposed that IVM can be used for this treatment [56,57] following the results of an in vitro study, which suggested that IVM may act against SARS-CoV-2 by blocking the nuclear import of viral proteins [57]. Since then, several recent systematic reviews [58,59,60] and other research reports [61,62] indicated the efficacy of ivermectin in the treatment and prevention of SARS-CoV-2 infection [58,63], while several other recent reports called for additional studies to clarify its use in COVID-19 treatment [64,65,66,67]. IVM has also demonstrated antiviral activity against other RNA and DNA viruses [68].

The proposed beneficial effect of IVM in the treatment of COVID-19 would accompany the enormous impact that this drug has already made in the treatment of a variety of parasitic infections that led to the Nobel Prize in 2015 and to its inclusion to the World Health Organization's Essential Medicines list [60,62,69]. IVM was found to be a highly effective, broad-spectrum, safe, and well tolerated drug [70], which, like other macrolide compounds, exhibited extremely wide diverse actions [71]. The inhibition of protease SARS-CoV-2 3CLProactivity that appears essential for viral replication [72,73] and the competitive binding of IVM with the viral S protein [74], were suggested as additional potential mechanisms that inhibit viral binding to ACE2 receptors and prevent consequent infection. In addition, it has been also reported that IVM has anti-inflammatory properties [75,76], which can interfere with the COVID-19 cytokine storm by inducing the activation of glycine receptors on leukocytes and vascular endothelium [77]. The results of a recent study demonstrated a high sensitivity of neuronal glycine receptors to IVM [78]. Thus, in line with the diverse biological functions of IVM, it would be important to further study its effects on glycine levels and whether regulation of glycine concentration perhaps includes the potential inhibition/silencing of the SIT1/SLC6A20 that may contribute to the treatment of COVID-19.

Regulation of glycine levels in Type 2 Diabetes

Recent metabolomic and genetic studies confirmed a strong association of T2D with decreased glycine levels in human subjects [79,80,81]. Furthermore, additional studies reported that plasma glycine concentration were decreased in obese and diabetic patients, and that the improved insulin sensitivity led to increased plasma glycine levels [82]. It appears that hypoglycinemia occurs even during the prediabetic state, however the effects of decreased glycine levels on T2D development are still not completely understood [82]. In addition, insulin sensitizer therapy with the combination of pioglitazone and metformin increased plasma glycine concentration as compared to the placebo treatment [83]. Furthermore, it was shown that IVM treatment enhanced the gene expression of two subunits of the glycine receptor, significantly modulated the mRNA expression of key markers in adipogenesis and fatty acid metabolism, as well as indicated an improved insulin sensitivity in vitro [84].

Conclusions

Recent studies demonstrated the association of SLC6A20 gene with the risk and severity of SARS-Cov-2 infection, where the preexisting hyperglycemia and diabetes diagnosis appear to be a strong predictor of mortality and adverse outcomes in COVID-19 patients. Previous reports also showed an association of SLC6A20 with Type 2 diabetes development. Based on these findings, SLC6A20/SIT1 may represent a novel target in the treatment of COVID-19, particularly in severe cases associated with T2D traits.

Furthermore, it was also recently proposed that SLC6A20 represents the novel regulator of glycine levels and that glycine has beneficial effects against the proinflammatory cytokine secretion induced by SARS-CoV-2 infection. It would be pertinent to further explore the possible mechanisms involved in the regulation of glycine levels, including the proposed inhibition of the SIT1 transporter and the activation of the glycine receptors by IVM or other potential drug candidates, which would result in increased levels of glycine. Hypoglycinemia has been found to be strongly associated with T2D, so it would be also important to analyze the glycine levels in COVID-19 patients, which seem not be studied yet. Therefore, additional clinical investigation is in order to confirm the potential favorable effects of targeting the SIT1 transporter and glycine levels for the efficient treatment of both, COVID-19 and Type 2 diabetes.

  1. Conflict of interest:

    The author declares no conflict of interest.

  2. Funding:

    The author received no specific funding for this work.

  3. Data availability statement:

    Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

References

1 Kasela S, Daniloski Z, Bollepalli S, Jordan TX, tenOever BR, Sanjana NE, et al. Integrative approach identifies SLC6A20 and CXCR6 as putative causal genes for the COVID-19 GWAS signal in the 3p21.31 locus. Genome Biol. 2021 Aug;22(1):242.10.1186/s13059-021-02454-4Search in Google Scholar PubMed PubMed Central

2 Yao Y, Ye F, Li K, Xu P, Tan W, Feng Q, et al. Genome and epigenome editing identify CCR9 and SLC6A20 as target genes at the 3p21.31 locus associated with severe COVID-19. Signal Transduct Target Ther. 2021 Feb;6(1):85.10.1038/s41392-021-00519-1Search in Google Scholar PubMed PubMed Central

3 Bae M, Roh JD, Kim Y, Kim SS, Han HM, Yang E, et al. SLC6A20 transporter: a novel regulator of brain glycine homeostasis and NMDAR function. EMBO Mol Med. 2021 Feb;13(2):e12632.10.15252/emmm.202012632Search in Google Scholar PubMed PubMed Central

4 Takanaga H, Mackenzie B, Suzuki Y, Hediger MA. Identification of mammalian proline transporter SIT1 (SLC6A20) with characteristics of classical system imino. J Biol Chem. 2005 Mar;280(10):8974–84.10.1074/jbc.M413027200Search in Google Scholar PubMed

5 Perlot T, Penninger JM. ACE2 - from the renin-angiotensin system to gut microbiota and malnutrition. Microbes Infect. 2013 Nov;15(13):866–73.10.1016/j.micinf.2013.08.003Search in Google Scholar PubMed PubMed Central

6 Vuille-dit-Bille RN, Camargo SM, Emmenegger L, Sasse T, Kummer E, Jando J, et al. Human intestine luminal ACE2 and amino acid transporter expression increased by ACE-inhibitors. Amino Acids. 2015 Apr;47(4):693–705.10.1007/s00726-014-1889-6Search in Google Scholar PubMed

7 Camargo SM, Vuille-Dit-Bille RN, Meier CF, Verrey F. ACE2 and gut amino acid transport. Clin Sci (Lond). 2020 Nov;134(21):2823–33.10.1042/CS20200477Search in Google Scholar PubMed

8 Ellinghaus D, Degenhardt F, Bujanda L, Buti M, Albillos A, Invernizzi P, et al.; Severe Covid-19 GWAS Group. Genomewide Association Study of Severe Covid-19 with Respiratory Failure. N Engl J Med. 2020 Oct;383(16):1522–34.10.1056/NEJMoa2020283Search in Google Scholar PubMed PubMed Central

9 Wang A, Chiou J, Poirion OB, Buchanan J, Valdez MJ, Verheyden JM, et al.; NHLBI LungMap Consortium. Single-cell multiomic profiling of human lungs reveals cell-type-specific and age-dynamic control of SARS-CoV2 host genes. eLife. 2020 Nov;9:9.10.7554/eLife.62522.sa2Search in Google Scholar

10 Camargo SM, Singer D, Makrides V, Huggel K, Pos KM, Wagner CA, et al. Tissue-specific amino acid transporter partners ACE2 and collectrin differentially interact with hartnup mutations. Gastroenterology. 2009 Mar;136(3):872–82.10.1053/j.gastro.2008.10.055Search in Google Scholar PubMed PubMed Central

11 Kaur U, Acharya K, Mondal R, Singh A, Saso L, Chakrabarti S, et al. Should ACE2 be given a chance in COVID-19 therapeutics: A semi-systematic review of strategies enhancing ACE2. Eur J Pharmacol. 2020 Nov;887:173545.10.1016/j.ejphar.2020.173545Search in Google Scholar PubMed PubMed Central

12 Erener S. Diabetes, infection risk and COVID-19. Mol Metab. 2020 Sep;39:101044.10.1016/j.molmet.2020.101044Search in Google Scholar PubMed PubMed Central

13 Guan WJ, Liang WH, Zhao Y, Liang HR, Chen ZS, Li YM, et al.; China Medical Treatment Expert Group for COVID-19. Comorbidity and its impact on 1590 patients with COVID-19 in China: a nationwide analysis. Eur Respir J. 2020 May;55(5):2000547.10.1183/13993003.00547-2020Search in Google Scholar PubMed PubMed Central

14 Richardson S, Hirsch JS, Narasimhan M, Crawford JM, McGinn T, Davidson KW, et al.; the Northwell COVID-19 Research Consortium. Presenting Characteristics, Comorbidities, and Outcomes Among 5700 Patients Hospitalized With COVID-19 in the New York City Area. JAMA. 2020 May;323(20):2052–9.10.1001/jama.2020.6775Search in Google Scholar PubMed PubMed Central

15 Shao F, Xu S, Ma X, Xu Z, Lyu J, Ng M, et al. In-hospital cardiac arrest outcomes among patients with COVID-19 pneumonia in Wuhan, China. Resuscitation. 2020 Jun;151:18–23.10.1016/j.resuscitation.2020.04.005Search in Google Scholar PubMed PubMed Central

16 Lim ZJ, Ponnapa Reddy M, Curtis JR, Afroz A, Billah B, Sheth V, et al. A Systematic Review of the Incidence and Outcomes of In-Hospital Cardiac Arrests in Patients With Coronavirus Disease 2019. Crit Care Med. 2021 Jun;49(6):901–11.10.1097/CCM.0000000000004950Search in Google Scholar PubMed

17 Whittaker A, Anson M, Harky A. Neurological Manifestations of COVID-19: A systematic review and current update. Acta Neurol Scand. 2020 Jul;142(1):14–22.10.1111/ane.13266Search in Google Scholar PubMed PubMed Central

18 O’Loughlin L, Alvarez Toledo N, Budrie L, Waechter R, Rayner J. A Systematic Review of Severe Neurological Manifestations in Pediatric Patients with Coexisting SARS-CoV-2 Infection. Neurol Int. 2021 Aug;13(3):410–27.10.3390/neurolint13030041Search in Google Scholar PubMed PubMed Central

19 Tufano A, Rendina D, Abate V, Casoria A, Marra A, Buonanno P, et al. Venous Thromboembolism in COVID-19 Compared to Non-COVID-19 Cohorts: A Systematic Review with Meta-Analysis. J Clin Med. 2021 Oct;10(21):4925.10.3390/jcm10214925Search in Google Scholar PubMed PubMed Central

20 Lim S, Bae JH, Kwon HS, Nauck MA. COVID-19 and diabetes mellitus: from pathophysiology to clinical management. Nat Rev Endocrinol. 2021 Jan;17(1):11–30.10.1038/s41574-020-00435-4Search in Google Scholar PubMed PubMed Central

21 Bonafè M, Prattichizzo F, Giuliani A, Storci G, Sabbatinelli J, Olivieri F. Inflamm-aging: why older men are the most susceptible to SARS-CoV-2 complicated outcomes. Cytokine Growth Factor Rev. 2020 Jun;53:33–7.10.1016/j.cytogfr.2020.04.005Search in Google Scholar

22 Prattichizzo F, Sabbatinelli J, de Candia P, Olivieri F, Ceriello A. Tackling the pillars of ageing to fight COVID-19. Lancet Healthy Longev. 2021 Apr;2(4):e191.10.1016/S2666-7568(21)00053-2Search in Google Scholar

23 Schlesinger S, Neuenschwander M, Lang A, Pafili K, Kuss O, Herder C, et al. Risk phenotypes of diabetes and association with COVID-19 severity and death: a living systematic review and meta-analysis. Diabetologia. 2021 Jul;64(7):1480–91.10.1007/s00125-021-05458-8Search in Google Scholar

24 Zhu Z, Mao Y, Chen G. Predictive value of HbA1c for in-hospital adverse prognosis in COVID-19: A systematic review and meta-analysis. Prim Care Diabetes. 2021 Aug;15(6):S1751-9918(21)00130-3. https://doi.org/10.1016/j.pcd.2021.07.013.10.1016/j.pcd.2021.07.013Search in Google Scholar

25 Yang Y, Cai Z, Zhang J. Hyperglycemia at admission is a strong predictor of mortality and severe/critical complications in COVID-19 patients: a meta-analysis. Biosci Rep. 2021 Feb;41(2):BSR20203584. https://doi.org/10.1042/BSR20203584.10.1042/BSR20203584Search in Google Scholar

26 Lazarus G, Audrey J, Wangsaputra VK, Tamara A, Tahapary DL. High admission blood glucose independently predicts poor prognosis in COVID-19 patients: A systematic review and dose-response meta-analysis. Diabetes Res Clin Pract. 2021 Jan;171:108561.10.1016/j.diabres.2020.108561Search in Google Scholar

27 Chen J, Wu C, Wang X, Yu J, Sun Z. The Impact of COVID-19 on Blood Glucose: A Systematic Review and Meta-Analysis. Front Endocrinol (Lausanne). 2020 Oct;11:574541.10.3389/fendo.2020.574541Search in Google Scholar

28 Prattichizzo F, de Candia P, Nicolucci A, Ceriello A. Elevated HbA1c levels in pre-Covid-19 infection increases the risk of mortality: A sistematic review and meta-analysis. Diabetes Metab Res Rev. 2021 May;•••:e3476.10.1002/dmrr.3476Search in Google Scholar

29 Ceriello A, Prattichizzo F. Pharmacological management of COVID-19 in type 2 diabetes. J Diabetes Complications. 2021 Jul;35(7):107927.10.1016/j.jdiacomp.2021.107927Search in Google Scholar

30 Bramante CT, Ingraham NE, Murray TA, Marmor S, Hovertsen S, Gronski J, et al. Metformin and risk of mortality in patients hospitalised with COVID-19: a retrospective cohort analysis. Lancet Healthy Longev. 2021 Jan;2(1):e34–41.10.1016/S2666-7568(20)30033-7Search in Google Scholar

31 Lalau JD, Al-Salameh A, Hadjadj S, Goronflot T, Wiernsperger N, Pichelin M, et al.; CORONADO investigators. Metformin use is associated with a reduced risk of mortality in patients with diabetes hospitalised for COVID-19. Diabetes Metab. 2021 Sep;47(5):101216.10.1016/j.diabet.2020.101216Search in Google Scholar PubMed PubMed Central

32 Li Y, Yang X, Yan P, Sun T, Zeng Z, Li S. Metformin in Patients With COVID-19: A Systematic Review and Meta-Analysis. Front Med (Lausanne). 2021 Aug;8:704666.10.3389/fmed.2021.704666Search in Google Scholar PubMed PubMed Central

33 Penlioglou T, Papachristou S, Papanas N. COVID-19 and Diabetes Mellitus: May Old Anti-diabetic Agents Become the New Philosopher's Stone? Diabetes Ther. 2020 May;11(6):1–3.10.1007/s13300-020-00830-0Search in Google Scholar PubMed PubMed Central

34 Bramante CT, Ingraham NE, Murray TA, Marmor S, Hovertsen S, Gronski J, et al. Observational Study of Metformin and Risk of Mortality in Patients Hospitalized with Covid-19. medRxiv. 2020. https://doi.org/10.1101/2020.06.19.20135095.10.1101/2020.06.19.20135095Search in Google Scholar PubMed PubMed Central

35 Sharma S, Ray A, Sadasivam B. Metformin in COVID-19: A possible role beyond diabetes. Diabetes Res Clin Pract. 2020 Jun;164:108183.10.1016/j.diabres.2020.108183Search in Google Scholar PubMed PubMed Central

36 Gheblawi M, Wang K, Viveiros A, Nguyen Q, Zhong JC, Turner AJ, et al. Angiotensin-Converting Enzyme 2: SARS-CoV-2 Receptor and Regulator of the Renin-Angiotensin System: Celebrating the 20th Anniversary of the Discovery of ACE2. Circ Res. 2020 May;126(10):1456–74.10.1161/CIRCRESAHA.120.317015Search in Google Scholar PubMed PubMed Central

37 Prattichizzo F, Giuliani A, Mensà E, Sabbatinelli J, De Nigris V, Rippo MR, et al. Pleiotropic effects of metformin: shaping the microbiome to manage type 2 diabetes and postpone ageing. Ageing Res Rev. 2018 Dec;48:87–98.10.1016/j.arr.2018.10.003Search in Google Scholar PubMed

38 Ling Y, van Herpt TT, van Hoek M, Dehghan A, Hofman A, Uitterlinden AG, et al. A genetic variant in SLC6A20 is associated with Type 2 diabetes in white-European and Chinese populations. Diabet Med. 2014 Nov;31(11):1350–6.10.1111/dme.12528Search in Google Scholar PubMed

39 Gemmati D, Tisato V. Genetic Hypothesis and Pharmacogenetics Side of Renin-Angiotensin-System in COVID-19. Genes (Basel). 2020 Sep;11(9):E1044.10.3390/genes11091044Search in Google Scholar PubMed PubMed Central

40 Abuissa H, Jones PG, Marso SP, O’Keefe JH Jr. Angiotensin-converting enzyme inhibitors or angiotensin receptor blockers for prevention of type 2 diabetes: a meta-analysis of randomized clinical trials. J Am Coll Cardiol. 2005 Sep;46(5):821–6.10.1016/j.jacc.2005.05.051Search in Google Scholar PubMed

41 Niklason A, Hedner T, Niskanen L, Lanke J; Captopril Prevention Project Study Group. Development of diabetes is retarded by ACE inhibition in hypertensive patients—a subanalysis of the Captopril Prevention Project (CAPPP). J Hypertens. 2004 Mar;22(3):645–52.10.1097/00004872-200403000-00029Search in Google Scholar PubMed

42 Yusuf S, Gerstein H, Hoogwerf B, Pogue J, Bosch J, Wolffenbuttel BH, et al.; HOPE Study Investigators. Ramipril and the development of diabetes. JAMA. 2001 Oct;286(15):1882–5.10.1001/jama.286.15.1882Search in Google Scholar PubMed

43 Cheng Q, Shah N, Bröer A, Fairweather S, Jiang Y, Schmoll D, et al. Identification of novel inhibitors of the amino acid transporter B0 AT1 (SLC6A19), a potential target to induce protein restriction and to treat type 2 diabetes. Br J Pharmacol. 2017 Mar;174(6):468–82.10.1111/bph.13711Search in Google Scholar PubMed PubMed Central

44 Patterson AD, Bonzo JA, Li F, Krausz KW, Eichler GS, Aslam S, et al. Metabolomics reveals attenuation of the SLC6A20 kidney transporter in nonhuman primate and mouse models of type 2 diabetes mellitus. J Biol Chem. 2011 Jun;286(22):19511–22.10.1074/jbc.M111.221739Search in Google Scholar PubMed PubMed Central

45 Reich HN, Oudit GY, Penninger JM, Scholey JW, Herzenberg AM. Decreased glomerular and tubular expression of ACE2 in patients with type 2 diabetes and kidney disease. Kidney Int. 2008 Dec;74(12):1610–6.10.1038/ki.2008.497Search in Google Scholar PubMed

46 Mizuiri S, Hemmi H, Arita M, Ohashi Y, Tanaka Y, Miyagi M, et al. Expression of ACE and ACE2 in individuals with diabetic kidney disease and healthy controls. Am J Kidney Dis. 2008 Apr;51(4):613–23.10.1053/j.ajkd.2007.11.022Search in Google Scholar PubMed

47 Pal R, Bhadada SK. COVID-19 and diabetes mellitus: an unholy interaction of two pandemics. Diabetes Metab Syndr. 2020 Jul–Aug;14(4):513–7.10.1016/j.dsx.2020.04.049Search in Google Scholar PubMed PubMed Central

48 Lu CL, Wang Y, Yuan L, Li Y, Li XY. The angiotensin-converting enzyme 2/angiotensin (1–7)/Mas axis protects the function of pancreatic β cells by improving the function of islet microvascular endothelial cells. Int J Mol Med. 2014 Nov;34(5):1293–300.10.3892/ijmm.2014.1917Search in Google Scholar PubMed

49 Cao X, Yang FY, Xin Z, Xie RR, Yang JK. The ACE2/Ang-(1–7)/Mas axis can inhibit hepatic insulin resistance. Mol Cell Endocrinol. 2014 Aug;393(1–2):30–8.10.1016/j.mce.2014.05.024Search in Google Scholar PubMed

50 Zhong JC, Yu XY, Lin QX, Li XH, Huang XZ, Xiao DZ, et al. Enhanced angiotensin converting enzyme 2 regulates the insulin/Akt signalling pathway by blockade of macrophage migration inhibitory factor expression. Br J Pharmacol. 2008 Jan;153(1):66–74.10.1038/sj.bjp.0707482Search in Google Scholar PubMed PubMed Central

51 Ilias I, Zabuliene L. Hyperglycemia and the novel Covid-19 infection: possible pathophysiologic mechanisms. Med Hypotheses. 2020 Jun;139:109699.10.1016/j.mehy.2020.109699Search in Google Scholar PubMed PubMed Central

52 Singer D, Camargo SM, Ramadan T, Schäfer M, Mariotta L, Herzog B, et al. Defective intestinal amino acid absorption in Ace2 null mice. Am J Physiol Gastrointest Liver Physiol. 2012 Sep;303(6):G686–95.10.1152/ajpgi.00140.2012Search in Google Scholar PubMed

53 Li CY. Can Glycine Mitigate COVID-19 Associated Tissue Damage and Cytokine Storm? Radiat Res. 2020 Sep;194(3):199–201.10.1667/RADE-20-00146.1Search in Google Scholar PubMed PubMed Central

54 Shan Q, Haddrill JL, Lynch JW. Ivermectin, an unconventional agonist of the glycine receptor chloride channel. J Biol Chem. 2001 Apr;276(16):12556–64.10.1074/jbc.M011264200Search in Google Scholar PubMed

55 Lynagh T, Webb TI, Dixon CL, Cromer BA, Lynch JW. Molecular determinants of ivermectin sensitivity at the glycine receptor chloride channel. J Biol Chem. 2011 Dec;286(51):43913–24.10.1074/jbc.M111.262634Search in Google Scholar PubMed PubMed Central

56 Chosidow O, Bernigaud C, Guillemot D, Giraudeau B, Lespine A, Changeux JP, et al. Ivermectin as a potential treatment for COVID-19? PLoS Negl Trop Dis. 2021 Jun;15(6):e0009446.10.1371/journal.pntd.0009446Search in Google Scholar PubMed PubMed Central

57 Caly L, Druce JD, Catton MG, Jans DA, Wagstaff KM. The FDA-approved drug ivermectin inhibits the replication of SARS-CoV-2 in vitro. Antiviral Res. 2020 Jun;178:104787.10.1016/j.antiviral.2020.104787Search in Google Scholar PubMed PubMed Central

58 Bryant A, Lawrie TA, Dowswell T, Fordham EJ, Mitchell S, Hill SR, et al. Ivermectin for Prevention and Treatment of COVID-19 Infection: A Systematic Review, Meta-analysis, and Trial Sequential Analysis to Inform Clinical Guidelines. Am J Ther. 2021 Jun;28(4):e434–60.10.1097/MJT.0000000000001402Search in Google Scholar PubMed PubMed Central

59 Analysis C. Ivermectin for COVID-19: real-time meta analysis of 64 studies. 2021.Search in Google Scholar

60 Kory P, Meduri GU, Varon J, Iglesias J, Marik PE. Review of the Emerging Evidence Demonstrating the Efficacy of Ivermectin in the Prophylaxis and Treatment of COVID-19. Am J Ther. 2021 Apr;28(3):e299–318.10.1097/MJT.0000000000001377Search in Google Scholar PubMed PubMed Central

61 Santin AD, Scheim DE, McCullough PA, Yagisawa M, Borody TJ. Ivermectin: a multifaceted drug of Nobel prize-honoured distinction with indicated efficacy against a new global scourge, COVID-19. New Microbes New Infect. 2021 Aug;43:100924.10.1016/j.nmni.2021.100924Search in Google Scholar PubMed PubMed Central

62 Yagisawa M. FPJ, Hanaki H., Omura S. . Global trends in clinical studies of ivermectin in COVID-19. Japanese J Antib. 2021;74(1).Search in Google Scholar

63 Cobos-Campos R, Apiñaniz A, Parraza N, Cordero J, García S, Orruño E. Potential use of ivermectin for the treatment and prophylaxis of SARS-CoV-2 infection. Curr Res Transl Med. 2021 Oct;69(4):103309.10.1016/j.retram.2021.103309Search in Google Scholar PubMed PubMed Central

64 Cruciani M, Pati I, Masiello F, Malena M, Pupella S, De Angelis V. Ivermectin for Prophylaxis and Treatment of COVID-19: A Systematic Review and Meta-Analysis. Diagnostics (Basel). 2021 Sep;11(9):1645.10.3390/diagnostics11091645Search in Google Scholar PubMed PubMed Central

65 Deng J, Zhou F, Ali S, Heybati K, Hou W, Huang E, et al. Efficacy and safety of ivermectin for the treatment of COVID-19: A systematic review and meta-analysis. QJM. 2021 Sep;hcab247. https://doi.org/10.1093/qjmed/hcab247.10.1093/qjmed/hcab247Search in Google Scholar PubMed PubMed Central

66 Hagiya H, Otsuka F. Ivermectin for Coronavirus Disease 2019: Yet to Be Well Evaluated Before Clinical Use. Clin Ther. 2021 Sep;43(9):1622–3.10.1016/j.clinthera.2021.07.007Search in Google Scholar PubMed PubMed Central

67 Popp M, Stegemann M, Metzendorf MI, Gould S, Kranke P, Meybohm P, et al. Ivermectin for preventing and treating COVID-19. Cochrane Database Syst Rev. 2021 Jul;7:CD015017.10.1002/14651858.CD015017Search in Google Scholar

68 Heidary F, Gharebaghi R. Ivermectin: a systematic review from antiviral effects to COVID-19 complementary regimen. J Antibiot (Tokyo). 2020 Sep;73(9):593–602.10.1038/s41429-020-0336-zSearch in Google Scholar PubMed PubMed Central

69 World Health Organization. Geneva S. 22nd Model List of Essential Medicines. 2021.Search in Google Scholar

70 Crump A, Ōmura S. Ivermectin, ‘wonder drug’ from Japan: the human use perspective. Proc Jpn Acad, Ser B, Phys Biol Sci. 2011;87(2):13–28.10.2183/pjab.87.13Search in Google Scholar PubMed PubMed Central

71 Omura S. Ivermectin: 25 years and still going strong. Int J Antimicrob Agents. 2008 Feb;31(2):91–8.10.1016/j.ijantimicag.2007.08.023Search in Google Scholar PubMed

72 Anand K, Ziebuhr J, Wadhwani P, Mesters JR, Hilgenfeld R. Coronavirus main proteinase (3CLpro) structure: basis for design of anti-SARS drugs. Science. 2003 Jun;300(5626):1763–7.10.1126/science.1085658Search in Google Scholar PubMed

73 Mody V, Ho J, Wills S, Mawri A, Lawson L, Ebert MC, et al. Identification of 3-chymotrypsin like protease (3CLPro) inhibitors as potential anti-SARS-CoV-2 agents. Commun Biol. 2021 Jan;4(1):93.10.1038/s42003-020-01577-xSearch in Google Scholar PubMed PubMed Central

74 Lehrer S, Rheinstein PH. Ivermectin Docks to the SARS-CoV-2 Spike Receptor-binding Domain Attached to ACE2. In Vivo. 2020 Sep–Oct;34(5):3023–6.10.21873/invivo.12134Search in Google Scholar PubMed PubMed Central

75 Kircik LH, Del Rosso JQ, Layton AM, Schauber J. Over 25 Years of Clinical Experience With Ivermectin: An Overview of Safety for an Increasing Number of Indications. J Drugs Dermatol. 2016 Mar;15(3):325–32.Search in Google Scholar

76 DiNicolantonio JJ, Barroso J, McCarty M. Ivermectin may be a clinically useful anti-inflammatory agent for late-stage COVID-19. Open Heart. 2020 Sep;7(2):e001350.10.1136/openhrt-2020-001350Search in Google Scholar PubMed PubMed Central

77 DiNicolantonio JJ, Barroso-Aranda J, McCarty MF. Anti-inflammatory activity of ivermectin in late-stage COVID-19 may reflect activation of systemic glycine receptors. Open Heart. 2021 Apr;8(1):e001655.10.1136/openhrt-2021-001655Search in Google Scholar PubMed PubMed Central

78 Bukanova JV, Solntseva EI, Kondratenko RV, Skrebitsky VG. Antiviral Drug Ivermectin at Nanomolar Concentrations Inhibits Glycine-Induced Chloride Current in Rat Hippocampal Neurons. Bull Exp Biol Med. 2021 Mar;170(5):649–53.10.1007/s10517-021-05125-3Search in Google Scholar PubMed PubMed Central

79 White PJ, McGarrah RW, Herman MA, Bain JR, Shah SH, Newgard CB. Insulin action, type 2 diabetes, and branched-chain amino acids: A two-way street. Mol Metab. 2021 Oct;52:101261.10.1016/j.molmet.2021.101261Search in Google Scholar PubMed PubMed Central

80 Felig P, Marliss E, Cahill GF Jr. Plasma amino acid levels and insulin secretion in obesity. N Engl J Med. 1969 Oct;281(15):811–6.10.1056/NEJM196910092811503Search in Google Scholar PubMed

81 Palmer ND, Stevens RD, Antinozzi PA, Anderson A, Bergman RN, Wagenknecht LE, et al. Metabolomic profile associated with insulin resistance and conversion to diabetes in the Insulin Resistance Atherosclerosis Study. J Clin Endocrinol Metab. 2015 Mar;100(3):E463–8.10.1210/jc.2014-2357Search in Google Scholar PubMed PubMed Central

82 Adeva-Andany M, Souto-Adeva G, Ameneiros-Rodríguez E, Fernández-Fernández C, Donapetry-García C, Domínguez-Montero A. Insulin resistance and glycine metabolism in humans. Amino Acids. 2018 Jan;50(1):11–27.10.1007/s00726-017-2508-0Search in Google Scholar PubMed

83 Irving BA, Carter RE, Soop M, Weymiller A, Syed H, Karakelides H, et al. Effect of insulin sensitizer therapy on amino acids and their metabolites. Metabolism. 2015 Jun;64(6):720–8.10.1016/j.metabol.2015.01.008Search in Google Scholar PubMed PubMed Central

84 Qi W, Clark JM, Suvorov A, Park Y. Ivermectin decreases triglyceride accumulation by inhibiting adipogenesis of 3T3-L1 preadipocytes. Food Chem Toxicol. 2019 Sep;131:110576.10.1016/j.fct.2019.110576Search in Google Scholar PubMed

85 Sachdeva S, Desai R, Gupta U, Prakash A, Jain A, Aggarwal A. Admission Hyperglycemia in Non-diabetics Predicts Mortality and Disease Severity in COVID-19: a Pooled Analysis and Meta-summary of Literature. SN Compr Clin Med. 2020 Oct;2(11):1–6.10.1007/s42399-020-00575-8Search in Google Scholar PubMed PubMed Central

86 Ilias I, Diamantopoulos A, Pratikaki M, Botoula E, Jahaj E, Athanasiou N, et al. Glycemia, Beta-Cell Function and Sensitivity to Insulin in Mildly to Critically Ill Covid-19 Patients. Medicina (Kaunas). 2021 Jan;57(1):68.10.3390/medicina57010068Search in Google Scholar PubMed PubMed Central

87 Jafar N, Edriss H, Nugent K. The Effect of Short-Term Hyperglycemia on the Innate Immune System. Am J Med Sci. 2016 Feb;351(2):201–11.10.1016/j.amjms.2015.11.011Search in Google Scholar PubMed

88 Shao S, Yang Q, Pan R, Yu X, Chen Y. Interaction of Severe Acute Respiratory Syndrome Coronavirus 2 and Diabetes. Front Endocrinol (Lausanne). 2021 Oct;12:731974.10.3389/fendo.2021.731974Search in Google Scholar PubMed PubMed Central

89 Teuwen LA, Geldhof V, Pasut A, Carmeliet P. COVID-19: the vasculature unleashed. Nat Rev Immunol. 2020 Jul;20(7):389–91.10.1038/s41577-020-0343-0Search in Google Scholar PubMed PubMed Central

90 Burgos-Morón E, Abad-Jiménez Z, Marañón AM, Iannantuoni F, Escribano-López I, López-Domènech S, et al. Relationship Between Oxidative Stress, ER Stress, and Inflammation in Type 2 Diabetes: The Battle Continues. J Clin Med. 2019 Sep;8(9):E1385.10.3390/jcm8091385Search in Google Scholar PubMed PubMed Central

91 Lumpuy-Castillo J, Lorenzo-Almorós A, Pello-Lázaro AM, Sánchez-Ferrer C, Egido J, Tuñón J, et al. Cardiovascular Damage in COVID-19: Therapeutic Approaches Targeting the Renin-Angiotensin-Aldosterone System. Int J Mol Sci. 2020 Sep;21(18):E6471.10.3390/ijms21186471Search in Google Scholar PubMed PubMed Central

92 Bălută M, Vintilă V, Vintilă M. Renin-Angiotensin-Aldosterone system inhibition in prevention of diabetes mellitus. Rom J Intern Med. 2004;42(2):277–88.Search in Google Scholar

Received: 2021-10-31
Accepted: 2021-11-29
Published Online: 2021-12-30

© 2021 Sabina Semiz, published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 12.5.2024 from https://www.degruyter.com/document/doi/10.1515/bmc-2021-0017/html
Scroll to top button