Next Article in Journal
How Important Are Genes to Achieve Longevity?
Next Article in Special Issue
Electroconvulsive Therapy in Psychiatric Disorders: A Narrative Review Exploring Neuroendocrine–Immune Therapeutic Mechanisms and Clinical Implications
Previous Article in Journal
A Novel Loss-of-Function SEMA3E Mutation in a Patient with Severe Intellectual Disability and Cognitive Regression
Previous Article in Special Issue
GILT Expression in Human Melanoma Cells Enhances Generation of Antigenic Peptides for HLA Class II-Mediated Immune Recognition
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Sodium-Glucose Co-Transporter 2 Inhibitors in the Regulation of Inflammatory Processes in Animal Models

by
Sandra Feijóo-Bandín
1,2,*,†,
Alana Aragón-Herrera
1,2,†,
Manuel Otero-Santiago
1,
Laura Anido-Varela
1,
Sandra Moraña-Fernández
1,3,
Estefanía Tarazón
2,4,
Esther Roselló-Lletí
2,4,
Manuel Portolés
2,4,
Oreste Gualillo
5,
José Ramón González-Juanatey
1,2 and
Francisca Lago
1,2
1
Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS) and Xerencia de Xestión Integrada de Santiago de Compostela (XXIS/SERGAS), 15706 Santiago de Compostela, Spain
2
Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Institute of Health Carlos III, 28029 Madrid, Spain
3
Cardiology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
4
Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, 46026 Valencia, Spain
5
Laboratory of Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases, Institute of Biomedical Research (IDIS) and Xerencia de Xestión Integrada de Santiago de Compostela (XXIS/SERGAS), 15706 Santiago de Compostela, Spain
*
Author to whom correspondence should be addressed.
These authors contributed equally to this study.
Int. J. Mol. Sci. 2022, 23(10), 5634; https://doi.org/10.3390/ijms23105634
Submission received: 29 April 2022 / Revised: 14 May 2022 / Accepted: 16 May 2022 / Published: 18 May 2022

Abstract

:
Sodium-glucose co-transporter 2 inhibitors, also known as gliflozins, were developed as a novel class of anti-diabetic agents that promote glycosuria through the prevention of glucose reabsorption in the proximal tubule by sodium-glucose co-transporter 2. Beyond the regulation of glucose homeostasis, they resulted as being effective in different clinical trials in patients with heart failure, showing a strong cardio-renal protective effect in diabetic, but also in non-diabetic patients, which highlights the possible existence of other mechanisms through which gliflozins could be exerting their action. So far, different gliflozins have been approved for their therapeutic use in T2DM, heart failure, and diabetic kidney disease in different countries, all of them being diseases that have in common a deregulation of the inflammatory process associated with the pathology, which perpetuates and worsens the disease. This inflammatory deregulation has been observed in many other diseases, which led the scientific community to have a growing interest in the understanding of the biological processes that lead to or control inflammation deregulation in order to be able to identify potential therapeutic targets that could revert this situation and contribute to the amelioration of the disease. In this line, recent studies showed that gliflozins also act as an anti-inflammatory drug, and have been proposed as a useful strategy to treat other diseases linked to inflammation in addition to cardio-renal diseases, such as diabetes, obesity, atherosclerosis, or non-alcoholic fatty liver disease. In this work, we will review recent studies regarding the role of the main sodium-glucose co-transporter 2 inhibitors in the control of inflammation.

1. Introduction

Since the discovery of phlorizin as a natural glucose-lowering molecule with anti-diabetic properties [1,2], many efforts have been made to develop new anti-diabetic drugs based on its chemical structure while improving its bioavailability, stability, and biological function [3,4].
Phlorizin (phloretin-2-’O-β-D-glucopyranoside), also referred to as phloridzin, is an O-glucoside of phloretin, a member of the dihydrochalcone family that is, in turn, a subclass of flavonoids [5]. It was originally isolated from the bark of the apple tree and proposed as a promising drug to treat malaria, fever, and infectious diseases due to its bitter taste, similar to extracts from the cinchona and willow tree, which had known antipyretic properties [6,7]. Many decades later, von Mering described for the first time its glucosuric effect and associated reduction of glycemia [8]. Phlorizin’s main pharmacological action is to block renal and intestinal glucose absorption through the non-selective inhibition of both sodium-glucose co-transporters 1 and 2 (SGLT1/2) to promote glucose excretion, which in turn reduces circulating glucose levels and improves insulin resistance [1]. More recently, other effects of phlorizin have been described using different in vivo and in vitro approaches, including cardioprotection, neuroprotection, the improvement of non-alcoholic fatty liver disease (NAFLD) and hepatic metabolism, body weight reduction, or the regulation of inflammation, fibrosis, and oxidative stress (Table 1).
Based on the structure of phlorizin, other synthetic SGLT2 inhibitors (SGLT2i) have been developed in the last decade as therapeutic agents to treat type-2 diabetes mellitus (T2DM), which have also been proved to have beneficial effects beyond glucose lowering [9]. However, despite most of them having been approved for their use in different countries, the exact mechanism through which they exert their actions besides glycosuria is not fully understood so far. Anyway, it seems clear that the development of SGLT2i has supposed a considerable improvement in the treatment of T2DM, heart failure, and diabetic renal disease [10].
Inflammation is a protective response of the immune system to engage extreme alterations in homeostasis due to infection, toxic compounds, damaged cells, or irradiation, in which cells in the innate immune system, adaptive immune system, and inflammatory mediators promote the removal of the harmful stimuli and the initiation of the healing process [11,12]. Although its activation is vital to health, when a homeostatic disturbance persists over time it can lead to non-resolving chronic inflammation which creates a vicious circle that perpetuates and worsens the pathological state, such as, for example, adipose tissue inflammation in obesity, lipotoxicity, or hyperglycemia [11]. The deregulation of the inflammatory response is a well-known factor associated with numerous diseases, among them T2DM, heart failure, or diabetic renal disease, which are the main diseases for which the use of SGLT2i has been approved so far [13]. An understanding of the mechanisms that regulate the deregulation of the inflammatory response in pathological conditions is crucial to be able to elaborate more accurate therapeutic approaches aimed at the modulation of inflammation. Indeed, inflammation has been suggested as a potential therapeutic target of SGLT2i [14]. Thus, in this work, we will make an overview of the main SGLT2i developed and discuss its potential use to treat inflammatory processes according to the main publications on the topic indexed in the databases PubMed, Scopus, and Web of Science.
Table 1. Main phlorizin’s effects described in the bibliography beyond glycosuria and the improvement of insulin resistance.
Table 1. Main phlorizin’s effects described in the bibliography beyond glycosuria and the improvement of insulin resistance.
EffectTissue/Cell TypeAnimal Model
Anti-inflammatoryBladderDiabetic Akita mice [15]
RAW 264.7 macrophages [16]-
3T3-L1 cells differentiated into adipocytes [17]-
Brain
Serum, liver, and cecum
Gastrointestinal tract
Brain
Plasma
Aged mice [18]
High-fat diet-fed mice [19]
Rats with irritable bowel syndrome [20]
Mice with Alzheimer’s disease [21]
Ovariectomized rats under inflammation conditions [22]
Body weight reduction-Diabetic db/db mice [23,24]
High-fat diet-fed mice [19,25]
Lipolysis3T3-L1 cells differentiated into adipocytes [16]-
Anti-oxidantBrain and liverAged mice [18]
PC12 cells [18]-
Skeletal muscleMice with exercise-induced fatigue [26]
HepG2 cells [27]-
Liver
Serum, liver, and cecum
Rats with hepatic fibrosis [28]
High-fat diet-fed mice [19]
RenoprotectiveKidneyDiabetic db/db mice [23]
HepatoprotectiveLiverDiabetic mice with NAFLD [29]
LiverRats with hepatic fibrosis [28]
LiverHigh-fat diet-fed mice [19]
Regulation of gut microbiota homeostasisGut microbiota
Gut microbiota
High-fat diet-fed mice [19,25]
Diabetic db/db mice [24]
Bone homeostasisMC3T3-E1 cells differentiated into osteoblasts and in bone
Bone
SAM mice [30]
Ovariectomized rats under inflammation conditions [22]
Improvement of cognitive functionBrain
Brain
Mice with Alzheimer’s disease [21]
Swiss mice [31]
Anti-cancerKYSE450 and KYSE30 cell
lines [32]
-
CardioprotectiveHeart
ACBRI 5118 cells [33]
HUVEC cells [34]
Heart
Diabetic db/db mice [35]
-
Guinea Pig [36]
RAW 264.7: a macrophage-like, Abelson leukemia virus-transformed cell line derived from BALB/c mice; 3T3-L1: fibroblast cell line isolated from the embryo of a mouse; db/db: monogenic, insulin-resistant model of T2DM due to a spontaneous mutation in the leptin receptor; PC12: cell line derived from a transplantable rat pheochromocytoma; HepG2: cell line exhibiting epithelial-like morphology that was isolated from a hepatocellular carcinoma of a 15-year-old, white, male youth with liver cancer; MC3T3-E1: osteoblastic cell line established from a C57BL/6 mouse calvaria; SAM: senescence-accelerated mice; KYSE450 and KYSE30: human esophageal cancer cells; ACBRI 5118: primary human cardiac fibroblast cells.

2. SGLT1/2 Biology

The human solute carrier family 5 (SLC5) is made up of 12 members, which mainly transport small molecules such as carbohydrates, vitamins, amino acids, and organic ions such as choline or short-chain fatty acids across cell membranes [37]. Among them, SGLT1 and SGLT2, encoded by the genes SLC5A1 and SLC5A2, are the most studied due to their known association with congenital glucose–galactose malabsorption and familial renal glycosuria, respectively, when they are defective [38,39], and to their recent role as therapeutic targets for treating T2DM.
SGLT1/2 are transmembrane proteins that bind sodium at the extracellular surface. This opens a gate to trap outside sugar and transport both molecules into the cell by flipping the co-transport, and then flipping back to the original conformation to start over a cycle that occurs ∼1000 times/s at a physiological temperature [40]. SGLT1 and SGLT2 have a different affinity and capacity for glucose transport, as well as different expression patterns (Table 2) [41].
SGLT1/2 mediate sodium-dependent sugar transport driven by the electrochemical sodium gradient created by the sodium/potassium-ATPase, which pumps sodium outside the cell into the bloodstream, creating a sodium gradient between the two sides of the membrane that allows SGLT1/2 to co-transport sodium and sugar from the extracellular side into the cytosol [44] (Figure 1).
SGLT1 is mainly expressed in the brush border of the small intestine, where it translocates one D-glucose/galactose molecule together with two sodium ions from the diet into mature enterocytes, being responsible for the intestinal absorption of glucose and galactose [45]. Moreover, SGLT1 also behaves as a water and urea channel, which is important for the absorption of water by passive transport in the small intestine [46,47]. SGLT2 is mostly located in the luminal membrane of the S1 and S2 segments of the renal proximal convoluted tubule, co-transporting one D-glucose molecule together with one sodium ion from the glomerular filtrate into the proximal tubule epithelial cells, and being responsible for the reabsorption of ~ 90% of the filtered glucose, while the remaining ~ 10% is reabsorbed by SGLT1 in the S3 segment, where there is less luminal glucose [43]. This exchange alters the glucose concentration inside the cell, being passively transported through the basolateral side of the cell via facilitative glucose transporters (GLUT) into the interstitial space and subsequently to the bloodstream (Figure 1) [43].
In T2DM the capacity to reabsorb renal-filtered glucose seems to be enhanced, which contributes to the increasing levels of glucose circulating in these patients and to the worsening of the disease [48,49]. Whether SGLT2 renal expression can be affected by T2DM remains controversial. Different studies using rodent animal models of T2DM have shown, on the one hand, an increased renal expression of SGLT2 [50,51,52,53,54], and on the other hand, a decreased expression [55,56] compared with non-diabetic animals. As well, in humans, lower [57] and higher [58,59] renal SGLT2 expressions in diabetic patients compared with non-diabetic patients have also been described. Despite the experimental differences among them, taken together, there are more studies in the bibliography supporting the increased expression levels of SGLT2 in diabetes than the contrary. Anyway, SGLT2 inhibition has been established as an effective therapeutic approach to treating T2DM and associated co-morbidities [10].

3. SGLT2 Inhibitors

Phlorizin was the first natural SGLT1/2 inhibitor discovered [8]. Despite its benefits improving insulin resistance [1] and diabetic complications such as nephropathy [23], non-alcoholic fatty liver disease [29], diabetic cardiomyopathy [35], or endothelial dysfunction [34], phlorizin had several drawbacks as a therapeutic agent, which included: (1) low oral bioavailability [60], (2) poor intestinal absorption and rapid clearance in urine [60], (3) low stability, being metabolized to phloretin in the small intestine by β-glucosidases [9], which inhibits GLUT1 [61] (responsible for glucose uptake in many tissues including the brain [62]), and (4) low solubility in water [63]. All of these observations, and the fact that mutations in the SGLT1 gene are associated with intestinal glucose–galactose malabsorption and associated fatal diarrhea [38,64], and that osmotic water flow through SGLT1 is blocked by phlorizin [65], prevented phlorizin from its use as an anti-diabetic drug. This led the scientific community to focus their efforts in the development of other glycoside-based molecules with a higher affinity for the inhibition of SGLT2 (SGLT2i), which could also overcome phlorizin pharmacokinetic limitations.
All synthetic SGLT2i are formed by a sugar head group linked to a long aromatic aglycone tail [9]. The sugar moiety interacts with the glucose binding site and the aglycone part binds to the extracellular vestibule of SGLT2 in the outward-open conformation, interrupting the transport cycle [66]. The first class of synthetic SGLT2i were O-glucosides, such as phlorizin, but they were synthesized as pro-drugs that need to be metabolized in the liver into their active form, avoiding intestinal β-glucosidases degradation [9]. T-1095 was the first synthetic O-glucoside to be developed, T-1095A being its active form [67]. T-1095A was able to inhibit SGLT2-dependent glucose renal reabsorption at the same time that it decreased GLUT2 expression in the kidney, and reduced blood glucose and HbA1c levels in diabetic animal models [67,68]. This finding paved the way for the development of a new class of oral anti-diabetics commonly known as gliflozins, most of them approved for their use in different countries (Table 2).
After T-1095, other O-glucosides were synthesized as pro-drugs, such as sergliflozin [69] or remogliflozin [70]. However, O-glucosides-SGLT2i were still metabolically unstable due to their recognition and cleavage by the β-glucosidases in circulation [9]. Meanwhile, C-glucosides (stable analogs of the corresponding O-glucosides) are metabolically more stable, showing a better oral bioavailability, and have a similar IC50 value for SLGT2 inhibition, which is translated into a lower dosage to achieve the same inhibitory effect (100-fold to 3000-fold) [9]. So T-1095 and sergliflozin were discontinued after phase II trials to be replaced for a new generation of C-glucosides, while remogliflozin remains as a low-cost SGLT2i in India despite having to be administrated twice daily [71]. Apart from this exception, all of the SGLT2i approved nowadays for the treatment of T2DM and, in some cases, its associated complications, are C-glucosides: dapagliflozin, canagliflozin, empagliflozin, ipragliflozin, luseogliflozin, tofogliflozin, and ertugliflozin [9]. Particularly, sotagliflozin has been approved in Europe for the treatment of T1DM [72] (Table 3). To note, although SGLT2i are not fully selective for SGLT2 co-transporters and inhibit SGLT1 to a different degree, due to the considerably higher affinity for SGLT2 than for SGLT1 they are commonly referred to as SGLT2i.
The SGLT2i mechanism of action is independent of insulin signaling/sensitivity and seems to overcome the commoner side effects of traditional anti-diabetic drugs, including hypoglycemia, weight gain, cardiovascular risk, gastrointestinal system problems, or even cancer risk [73].
Table 3. Main SGLT2i developed, their human SGLT2/1 selectivity calculated by IC50 (concentration causing half of the maximal inhibition) SGLT1/SGLT2 and first approved therapeutic use.
Table 3. Main SGLT2i developed, their human SGLT2/1 selectivity calculated by IC50 (concentration causing half of the maximal inhibition) SGLT1/SGLT2 and first approved therapeutic use.
Generic NameBrand NameCompanySGLT2 Selectivity over SGLT1First Global ApprovalTherapeutic Use
TofogliflozinApleway®Chugai Pharmaceutical Co. ~2912 fold [74]PMDA-2014 [75]T2DM [75]
EmpagliflozinJardiance®Boehringer Ingelheim GmbH. and Eli Lilly and Company ~2600 fold [76]FDA-2014 [77]
FDA-2016 [78]
FDA-2021 [79]
FDA-2022 [80]
T2DM [77]
CV death in T2DM [78]
HFrEF [79]
Heart failure [80]
Bexagliflozin-Theracos Inc.~2435 fold [81]Under clinical trials-
ErtugliflozinSteglatro™Merck & Co. and Pfizer Inc.~2200 fold [82]FDA-2017 [83]T2DM [83]
LuseogliflozinLusefi®Taisho Pharmaceutical Holdings Co.~1730 fold [84]PMDA-2014 [85]T2DM [85]
DapagliflozinForxiga® (E.U.)
Farxiga® (U.S.)
AstraZeneca and Bristol-Myers Squibb Co.~1200 fold [86]EMA-2012 [87]
FDA-2020 [88]
FDA-2021 [89]
T2DM [87]
HFrEF [88]
CKD [89]
# RemogliflozinRemozen™Glenmark Pharmaceuticals Ltd.~902 fold [70]CDSCO-2019 [90]T2DM [90]
# Sergliflozin-Glaxo Smith Kline (GSK) Plc.~300 fold [69]Discontinued after Phase II [43]-
IpragliflozinSuglat®Astellas Pharma Inc. and Kotobuki Pharmaceutical Co.~254 fold [91]PMDA -2014 [92]T2DM [92]
CanagliflozinInvokana®Janssen Global Services, L.L.C. and Mitsubishi Tanabe Pharma Co.~155 fold [93]FDA-2013 [94]
FDA-2018 [95]
FDA-2019 [96]
T2DM [94]
CV risk in T2DM [95]
DKD and risk of hospitalization for HF in T2DM [96]
# T-1095-Tanabe Seiyaku Company Ltd.~59 fold [76]Discontinued after Phase II [97]-
Licogliflozin-Novartis International A.G.~35 fold [98]Under clinical trials-
SotagliflozinZynquista™Sanofi-Aventis Group S.A. and Lexicon Pharmaceuticals, Inc.~20 fold [99]EMA-2019 [72]T1DM [72]
# Phlorizin--~13 fold [76]--
PMDA: Pharmaceuticals and Medical Devices Agency, Japan; FDA: U.S. Food and Drug Administration; CDSCO: Central Drugs Standard Control Organization, India; T2DM: type-2 diabetes mellitus; CV: cardiovascular; EMA: European Medicines Agency; HFrEF: heart failure with reduced ejection fraction; CKD: chronic kidney disease; DKD: diabetic kidney disease. # O-glucosides, the rest of the compounds are C-glucosides.
Moreover, apart from their direct benefit in reducing glycemia, the greater advantage of SGLT2i is that they have a cardio/renoprotective effect not only in diabetic, but also in non-diabetic patients, although the mechanism remains unclear [65,66,67,68].
Since T2DM is a heterogeneous disease with different associated co-morbidities, and the fact that T2DM patients´ response to the glucose-lowering agents available is quite variable [100], SGLT2i are currently approved for their use in combination with other drugs, complementing their effectiveness to achieve a personalized medicine depending on the clinical characteristics of patients, not only in T2DM, but also in cardio/renal diseases outside the context of T2DM [101,102].
Although the first interest in SGLT2i was their glucose-lowering capacity, lately there have been described pleiotropic effects of SGLT2i in a wide range of targets throughout the body (Figure 2). Using different experimental approaches and in different scenarios, SGLT2i have been shown to participate in the regulation of osmotic natriuresis and diuresis [103,104], hypertension [105], glucagon [106,107] and energy metabolism [108,109,110,111,112], mitochondrial function and biogenesis [113,114,115,116,117,118], autophagy [112,119,120,121,122,123,124,125], oxidative stress [46,123,126,127,128,129], fibrosis [46,130,131,132,133], apoptosis [122,128,134,135,136,137,138,139], endoplasmic reticulum stress [124,139,140,141,142,143], or inflammation [120,125,126,129,132,137,144,145], which opens a window to explore new uses of these drugs to treat other pathologies.

4. SGLT2i and Inflammation

On one hand, as mentioned in the introduction of this review, phlorizin, the first natural SGLT1/2 inhibitor discovered, is a molecule that has anti-inflammatory properties [146,147,148], along with other natural and synthetic chalcones and flavonoids [149,150,151]. On the other hand, the fact that inflammation and metabolism have a strong interplay and that their deregulation drives the development of several diseases is becoming increasingly evident. Normal cellular homeostasis relies on the crosstalk between the immune system and metabolic regulation in such a way that the pathogenesis of metabolic disorders is often triggered by or associated with inflammatory processes [152,153,154,155,156]. In keeping with this, many authors have started to refer to this fact as metaflammation [157,158].
Taking into account that synthetic SGLT2i are based on the chemical structure of phlorizin, the close interplay between inflammation and metabolic deregulation, and the positive effects of SGLT2i observed in pathologies with metabolic and inflammatory derangement, such as T2DM [159], heart failure [160], diabetic cardiomyopathy [161], diabetic nephropathy [162], or NAFLD [163], it was natural to explore their role as potential therapeutic agents to treat inflammation. Recently, several studies have described the anti-inflammatory effect of SGLT2i using different experimental approaches (Figure 3).

4.1. Regulation of Macrophage Tissue Infiltration, Polarization, and Cytokine Production

Macrophages are resident or infiltrated innate immune cells present in every tissue that are crucial to regulating not only the proper defense against pathogens, but also normal tissue homeostasis and repair [164]. They are characterized by an extraordinary plasticity that allows them to shift from one phenotype to another depending on the surrounding micro-environment (polarization), which also makes them active players that contribute to damage in pathological states of infection and inflammation [165]. According to this plasticity, macrophages are traditionally classified into two subgroups: M1 macrophages (classically activated), which are activated by Th1-type cytokines or bacterial lipopolysaccharides (LPS) and, in turn, produce pro-inflammatory cytokines, and M2 macrophages (alternatively activated), which are activated by Th2-type cytokines and have anti-inflammatory functions [165]. Although a short-term adaptive inflammatory response is necessary for defense and tissue repair, sustained inflammation is often harmful and contributes to disease progression [153]. However, once macrophages are polarized, they can be re-polarized back into M1 or M2 if they are exposed to the proper signals, a quality that has encouraged the scientific community to use them as a therapeutic target [165]. SGLT2i have been proved to be able to modulate M1/M2 macrophage polarization and infiltration in different conditions, with more studies having been carried out for dapagliflozin, canagliflozin, and empagliflozin.

4.1.1. Dapagliflozin

In macrophages obtained from healthy humans and polarized into M1 with LPS/interferon-gamma (INFγ) in culture, dapagliflozin pre-treatment can diminish the amount of M1 macrophages and increase the number of M2 macrophages, reducing the M1/M2 ratio in both normoglycemic and hyperglycemic environments. Moreover, dapagliflozin is able to block the LPS-induced secretion of the pro-inflammatory cytokines, interleukin (IL) 1β (IL-1β), IL-6, IL-8, and tumor necrosis factor-α (TNFα), as well as able to increase the expression of the toll-like receptor 4 (TLR4) and activate the nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) in macrophages [145], both of them important regulators of immune and inflammatory responses [166,167].
In the cardiovascular system, in infarcted rat hearts, dapagliflozin treatment after infarction increases myocardial M2 macrophage polarization with a concomitant decrease in M1 via acute-phase response factor (STAT3) signaling, a well-known key factor in the polarization of M2, contributing to attenuate cardiac fibrosis [168,169]. In apolipoprotein E-deficient (ApoE−/−) mice with T2DM induced with streptozotocin (STZ), dapagliflozin treatment can attenuate atherosclerosis lesions and decrease atherosclerotic macrophage infiltration [170,171], an effect also observed in ApoE−/− mice fed with a high-fat diet [172], in a normoglycemic rabbit model of atherosclerosis [173], in LDL-receptor-deficient (Ldlr−/−) mice with a high-fat and high-sucrose diabetogenic diet [174], or in T2DM rats [175]. Moreover, in mice with abdominal aortic aneurysms, dapagliflozin reduces the risk and progression of small aneurysms in part by attenuating aortic wall macrophage infiltration [176].
On the other hand, in diabetic ApoE−/− and db/db mice with hepatic steatosis, dapagliflozin reduces macrophage infiltration in the liver, contributing to the amelioration of liver fibrosis and inflammation [177,178]. Regarding the kidney, in T2DM db/db mice (mutation in the leptin receptor gene), diabetic otsuka long-evans tokushima fatty (OLETF) rats, and a type I diabetic Akita mouse model (AKITA/Slc) with diabetic nephropathy, dapagliflozin decreases macrophage infiltration and the reduction of the gene expression of the pro-inflammatory markers, monocyte chemoattractant protein-1 (MCP-1) and tumor growth factor-β (TGFβ) in the kidney [179,180,181], an effect also observed in mice with non-diabetic proteinuric nephropathy [182].

4.1.2. Canagliflozin

In C57BL/6 male mice with lung injury induced by LPS treatment, canagliflozin can inhibit M1 polarization and promote M2 polarization, reducing the M1/M2 macrophage ratio in the lung in vivo, and in bone marrow-derived macrophages in vitro, contributing to reduced inflammation [183]. As well as this, in a mouse model of human nonalcoholic steatohepatitis (NASH), canagliflozin shows benefits reducing liver inflammation and fibrosis by reducing macrophage infiltration and the M1/M2 macrophage ratio, and decreasing NFκB activity, alleviating acute heart injury [184]. In addition, in bone marrow-derived macrophages in culture stimulated with LPS, canagliflozin prevents the gene expression of IL-1β and TNFα, as well as TNFα secretion to the culture medium, associated with a decreased M1/M2 macrophages ratio [184].
In mice fed with a high-fat diet, canagliflozin suppresses the obesity-induced accumulation of macrophages along with a decrease in IL-6, and TNFα gene expression in the nodose ganglion and the hypothalamus, and a decrease of TNFα in skeletal muscle [185]. A similar effect is observed in ApoE−/− mice with atherosclerosis, where canagliflozin reduces the number of infiltrated macrophages in the atheroma plaque, which is related to the halting of the progression of the disease [186].
In diabetic New Zealand Obese (NZO, NZO/HlLtJ) mice and mice with T2DM, induced by treatment with nicotinamide and STZ, and by a high-sucrose diet, canagliflozin treatment reduces renal macrophage infiltration and fibrosis [187,188]. Moreover, in the macrophage THP-1 cell line in culture, canagliflozin activates AMP-activated protein kinase (AMPK) and inhibits the expression of the pro-inflammatory cytokines, IL-1β and IL-6, and monocyte chemoattractant protein-1 (MCP-1) [188]. Similarly, in cultured human endothelial cells, canagliflozin can hinder the IL-1β-stimulated secretion of IL-6 and MCP-1 in an AMPK-dependent manner [189].

4.1.3. Empagliflozin

Similar to dapagliflozin and canagliflozin, evidence of empagliflozin effects on macrophage polarization/infiltration in different experimental models exists. Empagliflozin has been shown to unbalance the amount of M1 macrophages in a culture of LPS-stimulated RAW 264.7 macrophages into M2 macrophages by inhibiting IκB kinase (IKK)/NFκB, mitogen-activated protein kinase kinase 7 (MKK7)/c-Jun N-terminal kinases (JNK), and janus kinase 2 (JAK2)/STAT1/3 pathways [190], and by activating AMPK [191].
In the cardiovascular system, empagliflozin reduces the amount of macrophage infiltration in the atheroma plaque and the perivascular adipose tissue in ApoE-/- mice, counteracting atherogenesis and vascular inflammation [192]. In this line, in STZ-diabetic mice, empagliflozin prevents the proliferation of plaque resident macrophages, which contributes to plaque regression [189]. On the other hand, in ApoE−/− mice with abdominal aortic aneurysms induced by angiotensin II, empagliflozin decreases macrophage infiltration in the aortic aneurysms sections [193]. Within the heart, empagliflozin shows early cardioprotective effects in diabetic db/db mice, which are in part due to the polarization of M1 macrophages into M2 in the cardiac tissue [194] and the reduction of cardiac macrophage infiltration [195]. In mice and rats with HFrEF, empagliflozin also reduces cardiac macrophage infiltration, contributing to the amelioration of the disease [196,197]. Furthermore, in C57BL/6J mice with corticosterone-induced cardiomyopathy, empagliflozin reduces M1 macrophage infiltration and increases the amount of M2 macrophages in the myocardium [198].
In C57BL/6J mice with T2DM and NAFLD induced by STZ and a high-fat diet, empagliflozin reduces the hepatic amount of M1 pro-inflammatory macrophages and induces autophagy in macrophages via activating the AMPK/mammalian target of the rapamycin (mTOR) signaling pathway and inhibiting the IL-17/IL-23 axis (which includes IL-1β, STAT3, or IL-6, among others), contributing to the amelioration of NAFLD [199]. Additionally, in C57BL/6J mice with NAFLD induced by choline-deficient, L-amino acid-defined, high-fat diet, empagliflozin attenuates M1 macrophage activation in the liver [200]. Moreover, in a mouse model of non-alcoholic steatohepatitis and diabetes, empagliflozin decreases the level of macrophage accumulation in the liver [201].
In mice and rats with diabetic nephropathy, empagliflozin can inhibit macrophage infiltration in the renal glomeruli, which is associated with decreased renal injury [202,203]. In line with this, empagliflozin also reduces macrophage infiltration in the kidney in male C57Bl/6N mice with LPS-induced renal injury [204], diabetic db/db mice [195], and in rats with renal fibrosis [205].
Regarding the adipose tissue, in ApoE−/− mice fed with a western diet, empagliflozin treatment reduces the amount of adipose tissue, the adipocyte size, and macrophage infiltration, contributing to reducing inflammation in the adipose tissue [206]. In diet-induced-obese mice, empagliflozin treatment alleviates obesity-induced inflammation and insulin resistance by decreasing M1-polarized macrophage accumulation, with the concomitant increase of the anti-inflammatory M2 phenotype, in fat and the liver, which, in part, promotes fat browning [207,208].

4.1.4. Other SGLT2i

In concordance with its family members dapagliflozin, canagliflozin, and empagliflozin, luseogliflozin and ipragliflozin also have proved anti-inflammatory effects. In diabetic ApoE−/− mice, luseogliflozin reduces the number of macrophages in the aorta, which correlates with the sizes of the atheroma areas [209], and ipragliflozin has been shown to have a similar effect in the femoral artery [210]. Moreover, ipragliflozin suppresses macrophage foam cell formation in mouse models of type 1 and T2DM and prevents atherosclerotic lesions along with dapagliflozin [171]. On the other hand, in C57BL/6 mice with STZ-induced diabetes, ipragliflozina ameliorates endothelial dysfunction, in part by reducing the amount of infiltrated macrophages in the abdominal aorta [211].
In the perivascular adipose tissue, both luseogliflozin and ipragliflozin decrease macrophage infiltration in mice fed with a high-fat diet and a western diet, respectively, contributing to counteracting adipose tissue remodeling [210,212]. On the other hand, ipragliflozin has been suggested to contribute to healthy adipose tissue expansion in high-fat diet-fed mice by reducing the M1/M2 macrophage ratio within the epididymal adipose tissue [213,214], and in the perirenal adipose tissue [215]. Furthermore, in high-fat diet-fed mice, ipragliflozin reduces the amount of macrophages in the liver and kidney [214,215].

4.2. Effects on NLRP3 Inflammasome

The NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome, the most widely studied inflammasome, is a multiprotein complex of the innate immune system that is mainly activated by pathogen infection and cellular damage. It is made up of a sensor (NLRP3, mainly expressed in macrophages and immune cells [216]), an adaptor (PYCARD, PYD, and CARD Domain Containing), and an effector (caspase 1), which cleaves and activates the pro-inflammatory cytokines pro-IL-1β and pro-IL-18, the end products of NLRP3 inflammasome activation that in turn enhance a pro-inflammatory response causing a cytokine storm [217,218]. On the other hand, caspase-1 activation can also cleavage and activate gasdermin D (GSDMD), the final effector of pyroptosis, a strong inflammatory process that leads to cell death in response to infection [219]. Although NRLP3 inflammasome activation is key to defense against pathogens and removing damaged cells, a deleterious activation of NLRP3 inflammasome and pyroptosis is associated with the pathogenesis of different inflammatory/metabolic diseases, including T2DM [220], cardiovascular disease [221], kidney disease [222], or liver disease [223], and its inhibition has been proved to be an effective therapeutic target [218,219].

4.2.1. Dapagliflozin

In diabetic ApoE−/− mice with NASH, dapagliflozin decreases the activation of the NLRP3 inflammasome in the liver, with the concomitant decrease in caspase-1 activity and secretion of IL-1β and IL-18 [177]. In accordance with this, also in ApoE−/− mice with diabetes and atherosclerosis, dapagliflozin treatment reduces circulating levels of NLRP3, IL-1β, and IL-18, as well as in the abdominal aorta, which contributes to reducing the atherosclerotic lesions and to increasing its stability [170]. Similarly, in mice with progressive T2DM (BTBR ob/ob), dapagliflozin decreases the myocardial expression of NLRP3, TNFα, and caspase-1, ameliorating the diabetic cardiomyopathy [224].
In rats with ulcerative colitis induced by acetic acid, dapagliflozin inhibits the NLRP3/caspase-1 signaling and decreases IL-1β expression in the colon through AMPK-dependent inactivation of NFκB [225]. The same effect is observed in rats subjected to chronic unpredictable stress, in which dapagliflozin reduces serum levels of both IL-1β and IL-18 cytokines, and decreases the expression of p-NFκB, NLRP3, caspase-1, IL-1β, and IL-18 in the hippocampus, suggesting a potential role of dapagliflozin in the treatment of depression [226].
In C57BL/6J mice with renal ischemia/reperfusion injury-induced kidney fibrosis, dapagliflozin decreases de expression of NLRP3 and blocks the activation of IL-1β, IL-18, and caspase-1 in the kidney [227]. In accordance with this, in BTBR ob/ob mice with diabetic nephropathy, dapagliflozin decreases the expression of NLRP3, TNFα, and caspase-1 in the kidney, along with an increase in AMPK phosphorylation, counteracting the disease progression [228].

4.2.2. Canagliflozin

In LPS-treated NIH Swiss male mice, canagliflozin inhibits NLRP3 inflammasome activation, shown by reduced plasma levels of IL-1β and IL-18, and by the reduced production of NLRP3, IL-1β, and IL-18 in the lung, an effect also observed in the mouse monocyte-macrophage cell line J774A.1 with LPS-stimulated NLRP3 activation, where the inactivation of NFκB and the promotion of AMPK phosphorylation induced by canagliflozin treatment seem to be key to blunt NLRP3 activation [229].

4.2.3. Empagliflozin

In T2DM patients at high risk of developing cardiovascular diseases, empagliflozin treatment can diminish NLRP3 inflammasome activation and secretion of IL-1β in isolated human macrophages after stimulation with LPS and Adenosine triphosphate (ATP) or palmitate, which could be one of the mechanisms that contribute to the proven empagliflozin´s cardioprotective effects [230]. Likewise, in human aortic smooth muscle cells in culture, empagliflozin can inhibit cell proliferation/migration, NLRP3 expression, caspase-1 activation, and the secretion of the pro-inflammatory markers IL-1β and IL-18 by targeting the signaling of the pro-inflammatory cytokine IL-17A, suggesting a protective role of empagliflozin on the pathogenesis of vascular proliferative diseases [231].
In mice with HFrEF, empagliflozin hinders NLRP3 activation by reducing the expression of NLRP3, NFκB, caspase-1, and TNFα, with the subsequent decrease of IL-1β and IL-18 expression in the myocardium, as well as in perfused hearts subjected to ischemia/reperfusion injury and in isolated human cardiomyocytes in culture stimulated with LPS [196]. On the other hand, in non-diabetic mice, empagliflozin protects the heart against doxorubicin cardiotoxicity by reducing the cardiac expression of NLRP3 and myeloid differentiation primary response 88 (MyD88, another central player of the innate immune response [232]), both of them recognized triggers of the cytokine storm involved in heart failure [233].
In mice with obesity and T2DM, the diet-induced activation of the NLRP3 inflammasome in the kidney and liver was blunted by empagliflozin treatment, shown by the reduction in caspase-1 activation and the production of IL-1β, along with lipid accumulation, an effect not observed in the heart despite the efficient reduction of cardiac lipid accumulation [234].
In pancreatic β TC-6 cells, an insulin-secreting cell line derived from transgenic mice expressing the large T-antigen of simian virus 40 (SV40) in pancreatic β-cells, empagliflozin treatment hinders the high-glucose-induced expression of NLRP3, caspase-1, and GSDMD, an effect also observed in vivo in the pancreas of diabetic mice that was associated with the prevention of pathological changes in pancreatic tissues [235]. In keeping with this, in db/db mice with a reduced ejection fraction and cardiac hypertrophy, empagliflozin also reduces NLRP3 and caspase-1 as well as the production of IL-1β and GSDMD, blocking cardiomyocyte pyroptosis in the diabetic heart [236].

5. Conclusions

The growing evidence linking the deregulation of inflammation with the triggering and maintenance of many diseases have opened new avenues for treatment by blocking this process. Understanding the mechanism through which this deregulation occurs is crucial for looking for effective therapeutic targets that could, at least, ameliorate the disease. SGLT2i have arrived in the clinical practice with strong evidence supporting their benefits, not only regarding glucose-lowering or cardiorenal outcomes but also in a wide range of tissues and pathologies. Taking into account the research carried out in recent years, it seems clear that SGLT2i have an effect protecting cells from inflammation, a process that could be involved in the pleiotropic beneficial effects of SGLT2i along with the improvement of energy metabolism, and that opens new lines of research regarding the potential role of SGLT2i as anti-inflammatory drugs.

Author Contributions

Conceptualization, S.F.-B., F.L. and J.R.G.-J.; writing—original draft preparation, S.F.-B. and A.A.-H.; writing—review and editing, S.F.-B., A.A.-H., M.O.-S., L.A.-V., S.M.-F., E.T., E.R.-L., M.P., R.L, O.G., J.R.G.-J. and F.L.; visualization, S.F.-B. and F.L.; supervision, F.L.; project administration, F.L.; funding acquisition, F.L. and J.R.G.-J. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Institute of Health “Fondo de Investigaciones Sanitarias del Instituto de Salud Carlos III” Madrid, Spain [PI21/01145, RETICS Program RD16/0012/0014 and CIBER de Enfermedades Cardiovasculares (CIBERCV)]; the European Regional Development Fund (FEDER) and European Union framework MSCA-RISE-H2020 Program (Project number 734899); Axencia Galega de Innovación (GAIN): Axudas do programa de consolidación e estruturación de unidades de investigación competitivas (GPC IN607B 2021 108). Sandra Moraña-Fernández was funded by predoctoral research grants from Xunta de Galicia, and Laura Anido-Varela was funded by the predoctoral research grant from the PFIS Program of the Spanish Ministry of Science and Instituto de Salud Carlos III (Spain).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rossetti, L.; Smith, D.; Shulman, G.I.; Papachristou, D.; DeFronzo, R.A. Correction of Hyperglycemia with Phlorizin Normalizes Tissue Sensitivity to Insulin in Diabetic Rats. J. Clin. Investig. 1987, 79, 1510–1515. [Google Scholar] [CrossRef] [PubMed]
  2. Chasis, H.; Jolliffe, N.; Smith, H.W. The Action of Phlorizin on the Excretion of Glucose, Xylose, Sucrose, Creatinine and Urea by Man. J. Clin. Investig. 1933, 12, 1083–1090. [Google Scholar] [CrossRef] [PubMed]
  3. Choi, C.-I. Sodium-Glucose Cotransporter 2 (SGLT2) Inhibitors from Natural Products: Discovery of Next-Generation Antihyperglycemic Agents. Molecules 2016, 21, 1136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. White, J.R. Apple Trees to Sodium Glucose Co-Transporter Inhibitors: A Review of SGLT2 Inhibition. Clin. Diabetes 2010, 28, 5–10. [Google Scholar] [CrossRef] [Green Version]
  5. Tian, L.; Cao, J.; Zhao, T.; Liu, Y.; Khan, A.; Cheng, G. The Bioavailability, Extraction, Biosynthesis and Distribution of Natural Dihydrochalcone: Phloridzin. Int. J. Mol. Sci. 2021, 22, 962. [Google Scholar] [CrossRef]
  6. Petersen, C. Analyse Des Phloridzins. Ann. Pharm. 1835, 15, 178. [Google Scholar] [CrossRef] [Green Version]
  7. Ehrenkranz, J.R.L.; Lewis, N.G.; Kahn, C.R.; Roth, J. Phlorizin: A Review. Diabetes Metab. Res. Rev. 2005, 21, 31–38. [Google Scholar] [CrossRef]
  8. Von Mering, J. Über Künstlichen Diabetes. Cent. Med. Wiss. 1886, 22, 31–35. [Google Scholar]
  9. Cai, W.; Jiang, L.; Xie, Y.; Liu, Y.; Liu, W.; Zhao, G. Design of SGLT2 Inhibitors for the Treatment of Type 2 Diabetes: A History Driven by Biology to Chemistry. Med. Chem. 2015, 11, 317–328. [Google Scholar] [CrossRef]
  10. Braunwald, E. SGLT2 Inhibitors: The Statins of the 21st Century. Eur. Heart J. 2022, 43, 1029–1030. [Google Scholar] [CrossRef]
  11. Kotas, M.E.; Medzhitov, R. Homeostasis, Inflammation, and Disease Susceptibility. Cell 2015, 160, 816–827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Libby, P. Inflammatory Mechanisms: The Molecular Basis of Inflammation and Disease. Nutr. Rev. 2007, 65, S140–S146. [Google Scholar] [CrossRef] [PubMed]
  13. Charlton, A.; Garzarella, J.; Jandeleit-Dahm, K.A.M.; Jha, J.C. Oxidative Stress and Inflammation in Renal and Cardiovascular Complications of Diabetes. Biology 2020, 10, 18. [Google Scholar] [CrossRef] [PubMed]
  14. Bonnet, F.; Scheen, A.J. Effects of SGLT2 Inhibitors on Systemic and Tissue Low-Grade Inflammation: The Potential Contribution to Diabetes Complications and Cardiovascular Disease. Diabetes Metab. 2018, 44, 457–464. [Google Scholar] [CrossRef] [PubMed]
  15. Inouye, B.M.; Hughes, F.M.; Jin, H.; Lütolf, R.; Potnis, K.C.; Routh, J.C.; Rouse, D.C.; Foo, W.C.; Todd Purves, J. Diabetic Bladder Dysfunction Is Associated with Bladder Inflammation Triggered through Hyperglycemia, not Polyuria. Res. Rep. Urol. 2018, 10, 219–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Huang, W.C.; Chang, W.T.; Wu, S.J.; Xu, P.Y.; Ting, N.C.; Liou, C.J. Phloretin and Phlorizin Promote Lipolysis and Inhibit Inflammation in Mouse 3T3-L1 Cells and in Macrophage-Adipocyte Co-Cultures. Mol. Nutr. Food Res. 2013, 57, 1803–1813. [Google Scholar] [CrossRef] [PubMed]
  17. Liddle, D.M.; Kavanagh, M.E.; Wright, A.J.; Robinson, L.E. Apple Flavonols Mitigate Adipocyte Inflammation and Promote Angiogenic Factors in LPS- and Cobalt Chloride-Stimulated Adipocytes, in Part by a Peroxisome Proliferator-Activated Receptor-γ-Dependent Mechanism. Nutrients 2020, 12, 1386. [Google Scholar] [CrossRef]
  18. Liu, Y.; Liu, Y.; Guo, Y.; Xu, L.; Wang, H. Phlorizin Exerts Potent Effects against Aging Induced by D-Galactose in Mice and PC12 Cells. Food Funct. 2021, 12, 2148–2160. [Google Scholar] [CrossRef]
  19. Liu, D.; Ji, Y.; Guo, Y.; Wang, H.; Wu, Z.; Li, H.; Wang, H. Dietary Supplementation of Apple Phlorizin Attenuates the Redox State Related to Gut Microbiota Homeostasis in C57BL/6J Mice Fed with a High-Fat Diet. J. Agric. Food Chem. 2021, 69, 198–211. [Google Scholar] [CrossRef]
  20. Nozu, T.; Miyagishi, S.; Ishioh, M.; Takakusaki, K.; Okumura, T. Phlorizin Attenuates Visceral Hypersensitivity and Colonic Hyperpermeability in a Rat Model of Irritable Bowel Syndrome. Biomed. Pharmacother. 2021, 139, 111649. [Google Scholar] [CrossRef]
  21. Rani, R.; Kumar, A.; Jaggi, A.S.; Singh, N. Pharmacological Investigations on Efficacy of Phlorizin a Sodium-Glucose Co-Transporter (SGLT) Inhibitor in Mouse Model of Intracerebroventricular Streptozotocin Induced Dementia of AD Type. J. Basic Clin. Physiol. Pharmacol. 2021, 32, 1057–1064. [Google Scholar] [CrossRef] [PubMed]
  22. Puel, C.; Quintin, A.; Mathey, J.; Obled, C.; Davicco, M.J.; Lebecque, P.; Kati-Coulibaly, S.; Horcajada, M.N.; Coxam, V. Prevention of Bone Loss by Phloridzin, an Apple Polyphenol, in Ovariectomized Rats under Inflammation Conditions. Calcif. Tissue Int. 2005, 77, 311–318. [Google Scholar] [CrossRef] [PubMed]
  23. Pei, F.; Li, B.-Y.Y.; Zhang, Z.; Yu, F.; Li, X.-L.L.; Lu, W.D.; Cai, Q.; Gao, H.-Q.Q.; Shen, L. Beneficial Effects of Phlorizin on Diabetic Nephropathy in Diabetic Db/Db Mice. J. Diabetes Complicat. 2016, 28, 596–603. [Google Scholar] [CrossRef] [PubMed]
  24. Mei, X.; Zhang, X.; Wang, Z.; Gao, Z.; Liu, G.; Hu, H.; Zou, L.; Li, X. Insulin Sensitivity-Enhancing Activity of Phlorizin Is Associated with Lipopolysaccharide Decrease and Gut Microbiota Changes in Obese and Type 2 Diabetes (Db/Db) Mice. J. Agric. Food Chem. 2016, 64, 7502–7511. [Google Scholar] [CrossRef] [PubMed]
  25. Zhang, X.Y.; Chen, J.; Yi, K.; Peng, L.; Xie, J.; Gou, X.; Peng, T.; Tang, L. Phlorizin Ameliorates Obesity-Associated Endotoxemia and Insulin Resistance in High-Fat Diet-Fed Mice by Targeting the Gut Microbiota and Intestinal Barrier Integrity. Gut Microbes 2020, 12, 1842990. [Google Scholar] [CrossRef] [PubMed]
  26. Ma, C.; Deng, Y.; Xiao, R.; Xu, F.; Li, M.; Gong, Q.; Gao, J. Anti-Fatigue Effect of Phlorizin on Exhaustive Exercise-Induced Oxidative Injury Mediated by Nrf2/ARE Signaling Pathway in Mice. Eur. J. Pharmacol. 2022, 918, 174563. [Google Scholar] [CrossRef]
  27. Wang, H.; Cheng, J.; Wang, H.; Wang, M.; Zhao, J.; Wu, Z. Protective Effect of Apple Phlorizin on Hydrogen Peroxide-Induced Cell Damage in HepG2 Cells. J. Food Biochem. 2019, 43, e13052. [Google Scholar] [CrossRef]
  28. Deng, G.; Wang, J.; Zhang, Q.; He, H.; Wu, F.; Feng, T.; Zhou, J.; Zou, K.; Hattori, M. Hepatoprotective Effects of Phloridzin on Hepatic Fibrosis Induced by Carbon Tetrachloride against Oxidative Stress-Triggered Damage and Fibrosis in Rats. Biol. Pharm. Bull. 2012, 35, 1118–1125. [Google Scholar] [CrossRef] [Green Version]
  29. David-Silva, A.; Esteves, J.V.; Morais, M.R.P.T.; Freitas, H.S.; Zorn, T.M.; Correa-Giannella, M.L.; Machado, U.F. Dual SGLT1/SGLT2 Inhibitor Phlorizin Ameliorates Non-Alcoholic Fatty Liver Disease and Hepatic Glucose Production in Type 2 Diabetic Mice. Diabetes. Metab. Syndr. Obes. 2020, 13, 739–751. [Google Scholar] [CrossRef] [Green Version]
  30. Antika, L.D.; Lee, E.-J.; Kim, Y.-H.; Kang, M.-K.; Park, S.-H.; Kim, D.Y.; Oh, H.; Choi, Y.-J.; Kang, Y.-H. Dietary Phlorizin Enhances Osteoblastogenic Bone Formation through Enhancing β-Catenin Activity via GSK-3β Inhibition in a Model of Senile Osteoporosis. J. Nutr. Biochem. 2017, 49, 42–52. [Google Scholar] [CrossRef]
  31. Boccia, M.M.; Kopf, S.R.; Baratti, C.M. Phlorizin, a Competitive Inhibitor of Glucose Transport, Facilitates Memory Storage in Mice. Neurobiol. Learn. Mem. 1999, 71, 104–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Jia, Z.; Xie, Y.; Wu, H.; Wang, Z.; Li, A.; Li, Z.; Yang, Z.; Zhang, Z.; Xing, Z.; Zhang, X. Phlorizin from Sweet Tea Inhibits the Progress of Esophageal Cancer by Antagonizing the JAK2/STAT3 Signaling Pathway. Oncol. Rep. 2021, 46, 137. [Google Scholar] [CrossRef] [PubMed]
  33. Meng, L.; Uzui, H.; Guo, H.; Tada, H. Role of SGLT1 in High Glucose Level-Induced MMP-2 Expression in Human Cardiac Fibroblasts. Mol. Med. Rep. 2018, 17, 6887–6892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Li, C.-Y.; Wang, L.-X.; Dong, S.-S.; Hong, Y.; Zhou, X.-H.; Zheng, W.-W.; Zheng, C. Phlorizin Exerts Direct Protective Effects on Palmitic Acid (PA)-Induced Endothelial Dysfunction by Activating the PI3K/AKT/ENOS Signaling Pathway and Increasing the Levels of Nitric Oxide (NO). Med. Sci. Monit. Basic Res. 2018, 24, 1–9. [Google Scholar] [CrossRef] [PubMed]
  35. Cai, Q.; Li, B.; Yu, F.; Lu, W.; Zhang, Z.; Yin, M.; Gao, H. Investigation of the Protective Effects of Phlorizin on Diabetic Cardiomyopathy in Db/Db Mice by Quantitative Proteomics. J. Diabetes Res. 2013, 2013, 263845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Hirose, M.; Shibazaki, T.; Nakada, T.; Kashihara, T.; Yano, S.; Okamoto, Y.; Isaji, M.; Matsushita, N.; Taira, E.; Yamada, M. Phlorizin Prevents Electrically-Induced Ventricular Tachyarrhythmia during Ischemia in Langendorff-Perfused Guinea-Pig Hearts. Biol. Pharm. Bull. 2014, 37, 1168–1176. [Google Scholar] [CrossRef] [Green Version]
  37. Gyimesi, G.; Pujol-Giménez, J.; Kanai, Y.; Hediger, M.A. Sodium-Coupled Glucose Transport, the SLC5 Family, and Therapeutically Relevant Inhibitors: From Molecular Discovery to Clinical Application. Pflug. Arch. 2020, 472, 1177–1206. [Google Scholar] [CrossRef]
  38. Lam, J.T.; Martín, M.G.; Turk, E.; Hirayama, B.A.; Bosshard, N.U.; Steinmann, B.; Wright, E.M. Missense Mutations in SGLT1 Cause Glucose–Galactose Malabsorption by Trafficking Defects. Biochim. Biophys. Acta-Mol. Basis Dis. 1999, 1453, 297–303. [Google Scholar] [CrossRef] [Green Version]
  39. Ottosson-Laakso, E.; Tuomi, T.; Forsén, B.; Gullström, M.; Groop, P.H.; Groop, L.; Vikman, P. Influence of Familial Renal Glycosuria Due to Mutations in the SLC5A2 Gene on Changes in Glucose Tolerance over Time. PLoS ONE 2016, 11, e0146114. [Google Scholar] [CrossRef]
  40. Ferrannini, E. Sodium-Glucose Co-Transporters and Their Inhibition: Clinical Physiology. Cell Metab. 2017, 26, 27–38. [Google Scholar] [CrossRef] [Green Version]
  41. DeFronzo, R.A.; Davidson, J.A.; Del Prato, S. The Role of the Kidneys in Glucose Homeostasis: A New Path towards Normalizing Glycaemia. Diabetes Obes. Metab. 2012, 14, 5–14. [Google Scholar] [CrossRef] [PubMed]
  42. Wright, E.M. Renal Na(+)-Glucose Cotransporters. Am. J. Physiol. Ren. Physiol. 2001, 280, F10–F18. [Google Scholar] [CrossRef] [PubMed]
  43. Abdul-Ghani, M.A.; Norton, L.; DeFronzo, R.A. Role of Sodium-Glucose Cotransporter 2 (SGLT 2) Inhibitors in the Treatment of Type 2 Diabetes. Endocr. Rev. 2011, 32, 515–531. [Google Scholar] [CrossRef] [Green Version]
  44. Isaji, M. SGLT2 Inhibitors: Molecular Design and Potential Differences in Effect. Kidney Int. Suppl. 2011, 1, S14–S19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Koepsell, H. Glucose Transporters in the Small Intestine in Health and Disease. Pflug. Arch. 2020, 472, 1207–1248. [Google Scholar] [CrossRef]
  46. Li, C.; Zhang, J.; Xue, M.; Li, X.; Han, F.; Liu, X.; Xu, L.; Lu, Y.; Cheng, Y.; Li, T.; et al. SGLT2 Inhibition with Empagliflozin Attenuates Myocardial Oxidative Stress and Fibrosis in Diabetic Mice Heart. Cardiovasc. Diabetol. 2019, 18, 15. [Google Scholar] [CrossRef] [PubMed]
  47. Loo, D.D.F.; Hirayama, B.A.; Meinild, A.K.; Chandy, G.; Zeuthen, T.; Wright, E.M. Passive Water and Ion Transport by Cotransporters. J. Physiol. 1999, 518, 195–202. [Google Scholar] [CrossRef]
  48. Vallon, V.; Thomson, S.C. Targeting Renal Glucose Reabsorption to Treat Hyperglycaemia: The Pleiotropic Effects of SGLT2 Inhibition. Diabetologia 2017, 60, 215–225. [Google Scholar] [CrossRef]
  49. DeFronzo, R.A.; Hompesch, M.; Kasichayanula, S.; Liu, X.; Hong, Y.; Pfister, M.; Morrow, L.A.; Leslie, B.R.; Boulton, D.W.; Ching, A.; et al. Characterization of Renal Glucose Reabsorption in Response to Dapagliflozin in Healthy Subjects and Subjects with Type 2 Diabetes. Diabetes Care 2013, 36, 3169–3176. [Google Scholar] [CrossRef] [Green Version]
  50. Osorio, H.; Bautista, R.; Rios, A.; Franco, M.; Arellano, A.; Vargas-Robles, H.; Romo, E.; Escalante, B. Effect of Phlorizin on SGLT2 Expression in the Kidney of Diabetic Rats. J. Nephrol. 2010, 23, 541–546. [Google Scholar]
  51. Osorio, H.; Bautista, R.; Rios, A.; Franco, M.; Santamaría, J.; Escalante, B. Effect of Treatment with Losartan on Salt Sensitivity and SGLT2 Expression in Hypertensive Diabetic Rats. Diabetes Res. Clin. Pract. 2009, 86, e46–e49. [Google Scholar] [CrossRef]
  52. Tabatabai, N.M.; Sharma, M.; Blumenthal, S.S.; Petering, D.H. Enhanced Expressions of Sodium–Glucose Cotransporters in the Kidneys of Diabetic Zucker Rats. Diabetes Res. Clin. Pract. 2009, 83, e27–e30. [Google Scholar] [CrossRef] [Green Version]
  53. Masuda, T.; Watanabe, Y.; Fukuda, K.; Watanabe, M.; Onishi, A.; Ohara, K.; Imai, T.; Koepsell, H.; Muto, S.; Vallon, V.; et al. Unmasking a Sustained Negative Effect of SGLT2 Inhibition on Body Fluid Volume in the Rat. Am. J. Physiol. Ren. Physiol. 2018, 315, F653–F664. [Google Scholar] [CrossRef] [PubMed]
  54. Hu, Z.; Liao, Y.; Wang, J.; Wen, X.; Shu, L. Potential Impacts of Diabetes Mellitus and Anti-Diabetes Agents on Expressions of Sodium-Glucose Transporters (SGLTs) in Mice. Endocrine 2021, 74, 571–581. [Google Scholar] [CrossRef] [PubMed]
  55. Albertoni Borghese, M.F.; Majowicz, M.P.; Ortiz, M.C.; del Passalacqua, M.R.; Sterin Speziale, N.B.; Vidal, N.A. Expression and Activity of SGLT2 in Diabetes Induced by Streptozotocin: Relationship with the Lipid Environment. Nephron. Physiol. 2009, 112, p45–p52. [Google Scholar] [CrossRef] [PubMed]
  56. Osorio, H.; Coronel, I.; Arellano, A.; Franco, M.; Escalante, B.; Bautista, R. Ursodeoxycholic Acid Decreases Sodium-Glucose Cotransporter (SGLT2) Expression and Oxidative Stress in the Kidney of Diabetic Rats. Diabetes Res. Clin. Pract. 2012, 97, 276–282. [Google Scholar] [CrossRef] [PubMed]
  57. Solini, A.; Rossi, C.; Mazzanti, C.M.; Proietti, A.; Koepsell, H.; Ferrannini, E. Sodium-Glucose Co-Transporter (SGLT)2 and SGLT1 Renal Expression in Patients with Type 2 Diabetes. Diabetes Obes. Metab. 2017, 19, 1289–1294. [Google Scholar] [CrossRef]
  58. Zhao, S.; Lo, C.-S.; Miyata, K.N.; Ghosh, A.; Zhao, X.-P.; Chenier, I.; Cailhier, J.-F.; Ethier, J.; Lattouf, J.-B.; Filep, J.G.; et al. Overexpression of Nrf2 in Renal Proximal Tubular Cells Stimulates Sodium-Glucose Cotransporter 2 Expression and Exacerbates Dysglycemia and Kidney Injury in Diabetic Mice. Diabetes 2021, 70, 1388–1403. [Google Scholar] [CrossRef]
  59. Rahmoune, H.; Thompson, P.W.; Ward, J.M.; Smith, C.D.; Hong, G.; Brown, J. Glucose Transporters in Human Renal Proximal Tubular Cells Isolated from the Urine of Patients with Non-Insulin-Dependent Diabetes. Diabetes 2005, 54, 3427–3434. [Google Scholar] [CrossRef] [Green Version]
  60. Crespy, V.; Aprikian, O.; Morand, C.; Besson, C.; Manach, C.; Demigné, C.; Rémésy, C. Bioavailability of Phloretin and Phloridzin in Rats. J. Nutr. 2001, 131, 3227–3230. [Google Scholar] [CrossRef] [Green Version]
  61. LeFevre, P.G. Sugar Transport in the Red Blood Cell: Structure-Activity Relationships in Substrates and Antagonists. Pharmacol. Rev. 1961, 13, 39–70. [Google Scholar] [PubMed]
  62. Maher, F.; Vannucci, S.J.; Simpson, I.A. Glucose Transporter Proteins in Brain. FASEB J. 1994, 8, 1003–1011. [Google Scholar] [CrossRef] [PubMed]
  63. Wang, Y.; Li, D.; Lin, H.; Jiang, S.; Han, L.; Hou, S.; Lin, S.; Cheng, Z.; Bian, W.; Zhang, X.; et al. Enhanced Oral Bioavailability and Bioefficacy of Phloretin Using Mixed Polymeric Modified Self-Nanoemulsions. Food Sci. Nutr. 2020, 8, 3545–3558. [Google Scholar] [CrossRef] [PubMed]
  64. Kasahara, M.; Maeda, M.; Hayashi, S.; Mori, Y.; Abe, T. A Missense Mutation in the Na+/Glucose Cotransporter Gene SGLT1 in a Patient with Congenital Glucose-Galactose Malabsorption: Normal Trafficking but Inactivation of the Mutant Protein. Biochim. Biophys. Acta-Mol. Basis Dis. 2001, 1536, 141–147. [Google Scholar] [CrossRef] [Green Version]
  65. Wright, E.M.; Ghezzi, C.; Loo, D.D.F. Novel and Unexpected Functions of SGLTs. Physiology 2017, 32, 435. [Google Scholar] [CrossRef]
  66. Bisignano, P.; Ghezzi, C.; Jo, H.; Polizzi, N.F.; Althoff, T.; Kalyanaraman, C.; Friemann, R.; Jacobson, M.P.; Wright, E.M.; Grabe, M. Inhibitor Binding Mode and Allosteric Regulation of Na+-Glucose Symporters. Nat. Commun. 2018, 9, 5245. [Google Scholar] [CrossRef]
  67. Oku, A.; Ueta, K.; Arakawa, K.; Ishihara, T.; Nawano, M.; Kuronuma, Y.; Matsumoto, M.; Saito, A.; Tsujihara, K.; Anai, M.; et al. T-1095, an Inhibitor of Renal Na+-Glucose Cotransporters, May Provide a Novel Approach to Treating Diabetes. Diabetes 1999, 48, 1794–1800. [Google Scholar] [CrossRef]
  68. Adachi, T.; Yasuda, K.; Okamoto, Y.; Shihara, N.; Oku, A.; Ueta, K.; Kitamura, K.; Saito, A.; Iwakura, I.; Yamada, Y.; et al. T-1095, a Renal Na+-Glucose Transporter Inhibitor, Improves Hyperglycemia in Streptozotocin-Induced Diabetic Rats. Metab.-Clin. Exp. 2000, 49, 990–995. [Google Scholar] [CrossRef]
  69. Katsuno, K.; Fujimori, Y.; Takemura, Y.; Hiratochi, M.; Itoh, F.; Komatsu, Y.; Fujikura, H.; Isaji, M. Sergliflozin, a Novel Selective Inhibitor of Low-Affinity Sodium Glucose Cotransporter (SGLT2), Validates the Critical Role of SGLT2 in Renal Glucose Reabsorption and Modulates Plasma Glucose Level. J. Pharmacol. Exp. Ther. 2007, 320, 323–330. [Google Scholar] [CrossRef] [Green Version]
  70. Shimizu, K.; Fujikura, H.; Fushimi, N.; Nishimura, T.; Tatani, K.; Katsuno, K.; Fujimori, Y.; Watanabe, S.; Hiratochi, M.; Nakabayashi, T.; et al. Discovery of Remogliflozin Etabonate: A Potent and Highly Selective SGLT2 Inhibitor. Bioorg. Med. Chem. 2021, 34, 116033. [Google Scholar] [CrossRef]
  71. Atal, S.; Fatima, Z.; Singh, S.; Balakrishnan, S.; Joshi, R. Remogliflozin: The New Low Cost SGLT-2 Inhibitor for Type 2 Diabetes Mellitus. Diabetol. Int. 2020, 12, 247–253. [Google Scholar] [CrossRef] [PubMed]
  72. European Medicines Agency Zynquista. 2019. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/zynquista (accessed on 11 April 2022).
  73. Osadebe, P.; Odoh, E.; Uzor, P. Natural Products as Potential Sources of Antidiabetic Drugs. Br. J. Pharm. Res. 2014, 4, 2075–2095. [Google Scholar] [CrossRef]
  74. Fediuk, D.J.; Nucci, G.; Dawra, V.K.; Cutler, D.L.; Amin, N.B.; Terra, S.G.; Boyd, R.A.; Krishna, R.; Sahasrabudhe, V. Overview of the Clinical Pharmacology of Ertugliflozin, a Novel Sodium-Glucose Cotransporter 2 (SGLT2) Inhibitor. Clin. Pharmacokinet. 2020, 59, 949–965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Poole, R.M.; Prossler, J.E. Tofogliflozin: First Global Approval. Drugs 2014, 74, 939–944. [Google Scholar] [CrossRef]
  76. Grempler, R.; Thomas, L.; Eckhardt, M.; Himmelsbach, F.; Sauer, A.; Sharp, D.E.; Bakker, R.A.; Mark, M.; Klein, T.; Eickelmann, P. Empagliflozin, a Novel Selective Sodium Glucose Cotransporter-2 (SGLT-2) Inhibitor: Characterisation and Comparison with Other SGLT-2 Inhibitors. Diabetes Obes. Metab. 2012, 14, 83–90. [Google Scholar] [CrossRef] [PubMed]
  77. Fala, L. Jardiance (Empagliflozin), an SGLT2 Inhibitor, Receives FDA Approval for the Treatment of Patients with Type 2 Diabetes. Am. Heal. Drug Benefits 2015, 8, 92–95. [Google Scholar]
  78. U.S. Food and Drug Administration. FDA Approves Jardiance to Reduce Cardiovascular Death in Adults with Type 2 Diabetes. 2016. Available online: https://www.fda.gov/news-events/press-announcements/fda-approves-jardiance-reduce-cardiovascular-death-adults-type-2-diabetes (accessed on 9 April 2022).
  79. Boehringer-Ingelheim US FDA Approves Jardiance® (Empagliflozin) to Treat Adults Living with Heart Failure with Reduced Ejection Fraction. 2021. Available online: https://www.boehringer-ingelheim.us/press-release/us-fda-approves-jardiance-empagliflozin-treat-adults-living-heart-failure-reduced (accessed on 9 April 2022).
  80. U.S. Food and Drug Administration. FDA Approves Treatment for Wider Range of Patients with Heart Failure. 2022. Available online: https://www.fda.gov/news-events/press-announcements/fda-approves-treatment-wider-range-patients-heart-failure (accessed on 9 April 2022).
  81. Azzam, O.; Carnagarin, R.; Lugo-Gavidia, L.M.; Nolde, J.; Matthews, V.B.; Schlaich, M.P. Bexagliflozin for Type 2 Diabetes: An Overview of the Data. Expert Opin. Pharmacother. 2021, 22, 2095–2103. [Google Scholar] [CrossRef]
  82. Mascitti, V.; Maurer, T.S.; Robinson, R.P.; Bian, J.; Boustany-Kari, C.M.; Brandt, T.; Collman, B.M.; Kalgutkar, A.S.; Klenotic, M.K.; Leininger, M.T.; et al. Discovery of a Clinical Candidate from the Structurally Unique Dioxa-Bicyclo[3.2.1]Octane Class of Sodium-Dependent Glucose Cotransporter 2 Inhibitors. J. Med. Chem. 2011, 54, 2952–2960. [Google Scholar] [CrossRef]
  83. Markham, A. Ertugliflozin: First Global Approval. Drugs 2018, 78, 513–519. [Google Scholar] [CrossRef]
  84. Kakinuma, H.; Oi, T.; Hashimoto-Tsuchiya, Y.; Arai, M.; Kawakita, Y.; Fukasawa, Y.; Iida, I.; Hagima, N.; Takeuchi, H.; Chino, Y.; et al. (1S)-1,5-Anhydro-1-[5-(4-Ethoxybenzyl)-2-Methoxy-4-Methylphenyl]-1-Thio-D-Glucitol (TS-071) Is a Potent, Selective Sodium-Dependent Glucose Cotransporter 2 (SGLT2) Inhibitor for Type 2 Diabetes Treatment. J. Med. Chem. 2010, 53, 3247–3261. [Google Scholar] [CrossRef]
  85. Markham, A.; Elkinson, S. Luseogliflozin: First Global Approval. Drugs 2014, 74, 945–950. [Google Scholar] [CrossRef] [PubMed]
  86. Meng, W.; Ellsworth, B.A.; Nirschl, A.A.; McCann, P.J.; Patel, M.; Girotra, R.N.; Wu, G.; Sher, P.M.; Morrison, E.P.; Biller, S.A.; et al. Discovery of Dapagliflozin: A Potent, Selective Renal Sodium-Dependent Glucose Cotransporter 2 (SGLT2) Inhibitor for the Treatment of Type 2 Diabetes. J. Med. Chem. 2008, 51, 1145–1149. [Google Scholar] [CrossRef] [PubMed]
  87. European Medicines Agency FORXIGATM (Dapagliflozin) Now Approved in European Union for Treatment of Type 2 Diabetes. 2014. Available online: https://www.astrazeneca.com/media-centre/press-releases/2012/FORXIGA-dapagliflozin-now-approved-in-European-Union-for-treatment-of-type-2-diabetes-14112012.html# (accessed on 11 April 2022).
  88. U.S. Food and Drug Administration. FDA Approves New Treatment for a Type of Heart Failure. 2020. Available online: https://www.fda.gov/news-events/press-announcements/fda-approves-new-treatment-type-heart-failure (accessed on 11 April 2022).
  89. U.S. Food and Drug Administration. FDA Approves Treatment for Chronic Kidney Disease. 2021. Available online: https://www.fda.gov/news-events/press-announcements/fda-approves-treatment-chronic-kidney-disease (accessed on 11 April 2022).
  90. Markham, A. Remogliflozin Etabonate: First Global Approval. Drugs 2019, 79, 1157–1161. [Google Scholar] [CrossRef] [PubMed]
  91. Takasu, T.; Yokono, M.; Tahara, A.; Takakura, S. In Vitro Pharmacological Profile of Ipragliflozin, a Sodium Glucose Co-Transporter 2 Inhibitor. Biol. Pharm. Bull. 2019, 42, 507–511. [Google Scholar] [CrossRef] [Green Version]
  92. Poole, R.M.; Dungo, R.T. Ipragliflozin: First Global Approval. Drugs 2014, 74, 611–617. [Google Scholar] [CrossRef]
  93. Liang, Y.; Arakawa, K.; Ueta, K.; Matsushita, Y.; Kuriyama, C.; Martin, T.; Du, F.; Liu, Y.; Xu, J.; Conway, B.; et al. Effect of Canagliflozin on Renal Threshold for Glucose, Glycemia, and Body Weight in Normal and Diabetic Animal Models. PLoS ONE 2012, 7, e30555. [Google Scholar] [CrossRef] [Green Version]
  94. Elkinson, S.; Scott, L.J. Canagliflozin: First Global Approval. Drugs 2013, 73, 979–988. [Google Scholar] [CrossRef]
  95. Johnson, J.; U.S. FDA. Approves INVOKANA® (Canagliflozin) to Reduce the Risk of Heart Attack, Stroke or Cardiovascular Death in Adults with Type 2 Diabetes and Established Cardiovascular Disease. 2018. Available online: https://www.jnj.com/u-s-fda-approves-invokana-canagliflozin-to-reduce-the-risk-of-heart-attack-stroke-or-cardiovascular-death-in-adults-with-type-2-diabetes-and-established-cardiovascular-disease (accessed on 11 April 2022).
  96. Johnson, J.; U.S. FDA. Approves INVOKANA® (Canagliflozin) to Treat Diabetic Kidney Disease (DKD) and Reduce the Risk of Hospitalization for Heart Failure in Patients with Type 2 Diabetes (T2D) and DKD. 2019. Available online: https://www.jnj.com/u-s-fda-approves-invokana-canagliflozin-to-treat-diabetic-kidney-disease-dkd-and-reduce-the-risk-of-hospitalization-for-heart-failure-in-patients-with-type-2-diabetes-t2d-and-dkd (accessed on 11 April 2022).
  97. Hardman, T.C.; Dubrey, S.W. Development and Potential Role of Type-2 Sodium-Glucose Transporter Inhibitors for Management of Type 2 Diabetes. Diabetes Ther. 2011, 2, 133–145. [Google Scholar] [CrossRef] [Green Version]
  98. Bays, H.E.; Kozlovski, P.; Shao, Q.; Proot, P.; Keefe, D. Licogliflozin, a Novel SGLT1 and 2 Inhibitor: Body Weight Effects in a Randomized Trial in Adults with Overweight or Obesity. Obesity 2020, 28, 870–881. [Google Scholar] [CrossRef] [Green Version]
  99. Cefalo, C.M.A.; Cinti, F.; Moffa, S.; Impronta, F.; Sorice, G.P.; Mezza, T.; Pontecorvi, A.; Giaccari, A. Sotagliflozin, the First Dual SGLT Inhibitor: Current Outlook and Perspectives. Cardiovasc. Diabetol. 2019, 18, 20. [Google Scholar] [CrossRef] [Green Version]
  100. Dennis, J.M. Precision Medicine in Type 2 Diabetes: Using Individualized Prediction Models to Optimize Selection of Treatment. Diabetes 2020, 69, 2075–2085. [Google Scholar] [CrossRef] [PubMed]
  101. Gloyn, A.L.; Drucker, D.J. Precision Medicine in the Management of Type 2 Diabetes. Lancet Diabetes Endocrinol. 2018, 6, 891–900. [Google Scholar] [CrossRef]
  102. Kalra, S.; Shetty, K.K.; Nagarajan, V.B.; Ved, J.K. Basic and Clinical Pharmaco-Therapeutics of SGLT2 Inhibitors: A Contemporary Update. Diabetes Ther. 2020, 11, 813–833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Yeoh, S.E.; Docherty, K.F.; Jhund, P.S.; Petrie, M.C.; Inzucchi, S.E.; Køber, L.; Kosiborod, M.N.; Martinez, F.A.; Ponikowski, P.; Sabatine, M.S.; et al. Relationship of Dapagliflozin With Serum Sodium. JACC Hear. Fail. 2022, 10, 306–318. [Google Scholar] [CrossRef]
  104. Masuda, T.; Muto, S.; Fukuda, K.; Watanabe, M.; Ohara, K.; Koepsell, H.; Vallon, V.; Nagata, D. Osmotic Diuresis by SGLT2 Inhibition Stimulates Vasopressin-Induced Water Reabsorption to Maintain Body Fluid Volume. Physiol. Rep. 2020, 8, e14360. [Google Scholar] [CrossRef] [Green Version]
  105. Upadhya, B.; Kozak, P.M.; Stacey, R.B.; Vasan, R.S. Newer Drugs to Reduce High Blood Pressure and Mitigate Hypertensive Target Organ Damage. Curr. Hypertens. Rep. 2022, 24, 1–20. [Google Scholar] [CrossRef]
  106. Saponaro, C.; Mühlemann, M.; Acosta-Montalvo, A.; Piron, A.; Gmyr, V.; Delalleau, N.; Moerman, E.; Thévenet, J.; Pasquetti, G.; Coddeville, A.; et al. Interindividual Heterogeneity of SGLT2 Expression and Function in Human Pancreatic Islets. Diabetes 2020, 69, 902–914. [Google Scholar] [CrossRef]
  107. Zhu, X.; Lin, C.; Li, L.; Hu, S.; Cai, X.; Ji, L. SGLT2i Increased the Plasma Fasting Glucagon Level in Patients with Diabetes: A Meta-Analysis. Eur. J. Pharmacol. 2021, 903, 174145. [Google Scholar] [CrossRef]
  108. Verma, S.; Rawat, S.; Ho, K.L.; Wagg, C.S.; Zhang, L.; Teoh, H.; Dyck, J.E.; Uddin, G.M.; Oudit, G.Y.; Mayoux, E.; et al. Empagliflozin Increases Cardiac Energy Production in Diabetes: Novel Translational Insights Into the Heart Failure Benefits of SGLT2 Inhibitors. JACC Basic Transl. Sci. 2018, 3, 575–587. [Google Scholar] [CrossRef]
  109. Marton, A.; Kaneko, T.; Kovalik, J.P.; Yasui, A.; Nishiyama, A.; Kitada, K.; Titze, J. Organ Protection by SGLT2 Inhibitors: Role of Metabolic Energy and Water Conservation. Nat. Rev. Nephrol. 2021, 17, 65–77. [Google Scholar] [CrossRef]
  110. Lee, J.Y.; Lee, M.; Lee, J.Y.; Bae, J.; Shin, E.; Lee, Y.H.; Lee, B.W.; Kang, E.S.; Cha, B.S. Ipragliflozin, an SGLT2 Inhibitor, Ameliorates High-Fat Diet-Induced Metabolic Changes by Upregulating Energy Expenditure through Activation of the AMPK/ SIRT1 Pathway. Diabetes Metab. J. 2021, 45, 921–932. [Google Scholar] [CrossRef] [PubMed]
  111. Wallenius, K.; Kroon, T.; Hagstedt, T.; Löfgren, L.; Sörhede-Winzell, M.; Boucher, J.; Lindén, D.; Oakes, N.D. The SGLT2 Inhibitor Dapagliflozin Promotes Systemic FFA Mobilization, Enhances Hepatic β-Oxidation, and Induces Ketosis. J. Lipid Res. 2022, 63, 100176. [Google Scholar] [CrossRef] [PubMed]
  112. Aragón-Herrera, A.; Feijóo-Bandín, S.; Otero Santiago, M.; Barral, L.; Campos-Toimil, M.; Gil-Longo, J.; Costa Pereira, T.M.; García-Caballero, T.; Rodríguez-Segade, S.; Rodríguez, J.; et al. Empagliflozin Reduces the Levels of CD36 and Cardiotoxic Lipids While Improving Autophagy in the Hearts of Zucker Diabetic Fatty Rats. Biochem. Pharmacol. 2019, 170, 113677. [Google Scholar] [CrossRef] [PubMed]
  113. Lee, Y.H.; Kim, S.H.; Kang, J.M.; Heo, J.H.; Kim, D.J.; Park, S.H.; Sung, M.J.; Kim, J.; Oh, J.; Yang, D.H.; et al. Empagliflozin Attenuates Diabetic Tubulopathy by Improving Mitochondrial Fragmentation and Autophagy. Am. J. Physiol.-Ren. Physiol. 2019, 317, F767–F780. [Google Scholar] [CrossRef] [PubMed]
  114. Mizuno, M.; Kuno, A.; Yano, T.; Miki, T.; Oshima, H.; Sato, T.; Nakata, K.; Kimura, Y.; Tanno, M.; Miura, T. Empagliflozin Normalizes the Size and Number of Mitochondria and Prevents Reduction in Mitochondrial Size after Myocardial Infarction in Diabetic Hearts. Physiol. Rep. 2018, 6, e13741. [Google Scholar] [CrossRef]
  115. Song, Y.; Huang, C.; Sin, J.; de Germano, J.F.; Taylor, D.J.R.; Thakur, R.; Gottlieb, R.A.; Mentzer, R.M.; Andres, A.M. Attenuation of Adverse Postinfarction Left Ventricular Remodeling with Empagliflozin Enhances Mitochondria-Linked Cellular Energetics and Mitochondrial Biogenesis. Int. J. Mol. Sci. 2021, 23, 437. [Google Scholar] [CrossRef]
  116. Belosludtsev, K.N.; Starinets, V.S.; Belosludtsev, M.N.; Mikheeva, I.B.; Dubinin, M.V.; Belosludtseva, N.V. Chronic Treatment with Dapagliflozin Protects against Mitochondrial Dysfunction in the Liver of C57BL/6NCrl Mice with High-Fat Diet/Streptozotocin-Induced Diabetes Mellitus. Mitochondrion 2021, 59, 246–254. [Google Scholar] [CrossRef]
  117. Wei, D.; Liao, L.; Wang, H.; Zhang, W.; Wang, T.; Xu, Z. Canagliflozin Ameliorates Obesity by Improving Mitochondrial Function and Fatty Acid Oxidation via PPARα In Vivo and In Vitro. Life Sci. 2020, 247, 117414. [Google Scholar] [CrossRef]
  118. Croteau, D.; Luptak, I.; Chambers, J.M.; Hobai, I.; Panagia, M.; Pimentel, D.R.; Siwik, D.A.; Qin, F.; Colucci, W.S. Effects of Sodium-Glucose Linked Transporter 2 Inhibition With Ertugliflozin on Mitochondrial Function, Energetics, and Metabolic Gene Expression in the Presence and Absence of Diabetes Mellitus in Mice. J. Am. Heart Assoc. 2021, 10, e019995. [Google Scholar] [CrossRef]
  119. Yang, L.; Liang, B.; Li, J.; Zhang, X.; Chen, H.; Sun, J.; Zhang, Z. Dapagliflozin Alleviates Advanced Glycation End Product Induced Podocyte Injury through AMPK/MTOR Mediated Autophagy Pathway. Cell. Signal. 2022, 90, 110206. [Google Scholar] [CrossRef]
  120. Xu, J.; Kitada, M.; Ogura, Y.; Liu, H.; Koya, D. Dapagliflozin Restores Impaired Autophagy and Suppresses Inflammation in High Glucose-Treated HK-2 Cells. Cells 2021, 10, 1457. [Google Scholar] [CrossRef] [PubMed]
  121. Li, L.; Li, Q.; Huang, W.; Han, Y.; Tan, H.; An, M.; Xiang, Q.; Zhou, R.; Yang, L.; Cheng, Y. Dapagliflozin Alleviates Hepatic Steatosis by Restoring Autophagy via the AMPK-MTOR Pathway. Front. Pharmacol. 2021, 12, 1190. [Google Scholar] [CrossRef] [PubMed]
  122. Arab, H.H.; Al-Shorbagy, M.Y.; Saad, M.A. Activation of Autophagy and Suppression of Apoptosis by Dapagliflozin Attenuates Experimental Inflammatory Bowel Disease in Rats: Targeting AMPK/MTOR, HMGB1/RAGE and Nrf2/HO-1 Pathways. Chem. Biol. Interact. 2021, 335, 109368. [Google Scholar] [CrossRef]
  123. Ala, M.; Khoshdel, M.R.F.; Dehpour, A.R. Empagliflozin Enhances Autophagy, Mitochondrial Biogenesis, and Antioxidant Defense and Ameliorates Renal Ischemia/Reperfusion in Nondiabetic Rats. Oxid. Med. Cell. Longev. 2022, 2022, 1197061. [Google Scholar] [CrossRef] [PubMed]
  124. Abdel-Rafei, M.; Thabet, N.; Rashed, L.; Moustafa, E. Canagliflozin, a SGLT-2 Inhibitor, Relieves ER Stress, Modulates Autophagy and Induces Apoptosis in Irradiated HepG2 Cells: Signal Transduction between PI3K/AKT/GSK-3β/MTOR and Wnt/β-Catenin Pathways; In Vitro. J. Cancer Res. Ther. 2021, 17, 1404–1418. [Google Scholar] [CrossRef] [PubMed]
  125. Xu, C.; Wang, W.; Zhong, J.; Lei, F.; Xu, N.; Zhang, Y.; Xie, W. Canagliflozin Exerts Anti-Inflammatory Effects by Inhibiting Intracellular Glucose Metabolism and Promoting Autophagy in Immune Cells. Biochem. Pharmacol. 2018, 152, 45–59. [Google Scholar] [CrossRef]
  126. Ashrafi Jigheh, Z.; Ghorbani Haghjo, A.; Argani, H.; Roshangar, L.; Rashtchizadeh, N.; Sanajou, D.; Nazari Soltan Ahmad, S.; Rashedi, J.; Dastmalchi, S.; Mesgari Abbasi, M. Empagliflozin Alleviates Renal Inflammation and Oxidative Stress in Streptozotocin-Induced Diabetic Rats Partly by Repressing HMGB1-TLR4 Receptor Axis. Iran. J. Basic Med. Sci. 2019, 22, 384–390. [Google Scholar] [CrossRef]
  127. Uthman, L.; Li, X.; Baartscheer, A.; Schumacher, C.A.; Baumgart, P.; Hermanides, J.; Preckel, B.; Hollmann, M.W.; Coronel, R.; Zuurbier, C.J.; et al. Empagliflozin Reduces Oxidative Stress through Inhibition of the Novel Inflammation/NHE/[Na+]c/ROS-Pathway in Human Endothelial Cells. Biomed. Pharmacother. 2022, 146, 112515. [Google Scholar] [CrossRef]
  128. Dasari, D.; Bhat, A.; Mangali, S.; Ghatage, T.; Lahane, G.P.; Sriram, D.; Dhar, A. Canagliflozin and Dapagliflozin Attenuate Glucolipotoxicity-Induced Oxidative Stress and Apoptosis in Cardiomyocytes via Inhibition of Sodium-Glucose Cotransporter-1. ACS Pharmacol. Transl. Sci. 2022, 5, 216–225. [Google Scholar] [CrossRef]
  129. Hazem, R.M.; Ibrahim, A.Z.; Ali, D.A.; Moustafa, Y.M. Dapagliflozin Improves Steatohepatitis in Diabetic Rats via Inhibition of Oxidative Stress and Inflammation. Int. Immunopharmacol. 2022, 104, 108503. [Google Scholar] [CrossRef]
  130. Chehrehgosha, H.; Sohrabi, M.R.; Ismail-Beigi, F.; Malek, M.; Reza Babaei, M.; Zamani, F.; Ajdarkosh, H.; Khoonsari, M.; Fallah, A.E.; Khamseh, M.E. Empagliflozin Improves Liver Steatosis and Fibrosis in Patients with Non-Alcoholic Fatty Liver Disease and Type 2 Diabetes: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Diabetes Ther. 2021, 12, 843–861. [Google Scholar] [CrossRef] [PubMed]
  131. Hayashizaki-Someya, Y.; Kurosaki, E.; Takasu, T.; Mitori, H.; Yamazaki, S.; Koide, K.; Takakura, S. Ipragliflozin, an SGLT2 Inhibitor, Exhibits a Prophylactic Effect on Hepatic Steatosis and Fibrosis Induced by Choline-Deficient l-Amino Acid-Defined Diet in Rats. Eur. J. Pharmacol. 2015, 754, 19–24. [Google Scholar] [CrossRef] [PubMed]
  132. Heerspink, H.J.L.; Perco, P.; Mulder, S.; Leierer, J.; Hansen, M.K.; Heinzel, A.; Mayer, G. Canagliflozin Reduces Inflammation and Fibrosis Biomarkers: A Potential Mechanism of Action for Beneficial Effects of SGLT2 Inhibitors in Diabetic Kidney Disease. Diabetologia 2019, 62, 1154–1166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Tian, J.; Zhang, M.; Suo, M.; Liu, D.; Wang, X.; Liu, M.; Pan, J.; Jin, T.; An, F. Dapagliflozin Alleviates Cardiac Fibrosis through Suppressing EndMT and Fibroblast Activation via AMPKα/TGF-β/Smad Signalling in Type 2 Diabetic Rats. J. Cell. Mol. Med. 2021, 25, 7642–7659. [Google Scholar] [CrossRef]
  134. Abdel-latif, R.G.; Rifaai, R.A.; Amin, E.F. Empagliflozin Alleviates Neuronal Apoptosis Induced by Cerebral Ischemia/Reperfusion Injury through HIF-1α/VEGF Signaling Pathway. Arch. Pharm. Res. 2020, 43, 514–525. [Google Scholar] [CrossRef]
  135. Tian, G.; Yu, Y.; Deng, H.; Yang, L.; Shi, X.; Yu, B. Empagliflozin Alleviates Ethanol-Induced Cardiomyocyte Injury through Inhibition of Mitochondrial Apoptosis via a SIRT1/PTEN/Akt Pathway. Clin. Exp. Pharmacol. Physiol. 2021, 48, 837–845. [Google Scholar] [CrossRef]
  136. Nakao, M.; Shimizu, I.; Katsuumi, G.; Yoshida, Y.; Suda, M.; Hayashi, Y.; Ikegami, R.; Hsiao, Y.T.; Okuda, S.; Soga, T.; et al. Empagliflozin Maintains Capillarization and Improves Cardiac Function in a Murine Model of Left Ventricular Pressure Overload. Sci. Rep. 2021, 11, 18384. [Google Scholar] [CrossRef]
  137. Faridvand, Y.; Kazemzadeh, H.; Vahedian, V.; Mirzajanzadeh, P.; Nejabati, H.R.; Safaie, N.; Maroufi, N.F.; Pezeshkian, M.; Nouri, M.; Jodati, A. Dapagliflozin Attenuates High Glucose-Induced Endothelial Cell Apoptosis and Inflammation through AMPK/SIRT1 Activation. Clin. Exp. Pharmacol. Physiol. 2022. [Google Scholar] [CrossRef]
  138. Hu, Y.; Xu, Q.; Li, H.; Meng, Z.; Hao, M.; Ma, X.; Lin, W.; Kuang, H. Dapagliflozin Reduces Apoptosis of Diabetic Retina and Human Retinal Microvascular Endothelial Cells Through ERK1/2/CPLA2/AA/ROS Pathway Independent of Hypoglycemic. Front. Pharmacol. 2022, 13, 497. [Google Scholar] [CrossRef]
  139. Shibusawa, R.; Yamada, E.; Okada, S.; Nakajima, Y.; Bastie, C.C.; Maeshima, A.; Kaira, K.; Yamada, M. Dapagliflozin Rescues Endoplasmic Reticulum Stress-Mediated Cell Death. Sci. Rep. 2019, 9, 9887. [Google Scholar] [CrossRef]
  140. Wang, C.-C.; Li, Y.; Qian, X.-Q.; Zhao, H.; Wang, D.; Zuo, G.-X.; Wang, K. Empagliflozin Alleviates Myocardial I/R Injury and Cardiomyocyte Apoptosis via Inhibiting ER Stress-Induced Autophagy and the PERK/ATF4/Beclin1 Pathway. J. Drug Target. 2022, 1–41. [Google Scholar] [CrossRef] [PubMed]
  141. Nasiri-Ansari, N.; Nikolopoulou, C.; Papoutsi, K.; Kyrou, I.; Mantzoros, C.S.; Kyriakopoulos, G.; Chatzigeorgiou, A.; Kalotychou, V.; Randeva, M.S.; Chatha, K.; et al. Empagliflozin Attenuates Non-Alcoholic Fatty Liver Disease (NAFLD) in High Fat Diet Fed ApoE(−/−) Mice by Activating Autophagy and Reducing ER Stress and Apoptosis. Int. J. Mol. Sci. 2021, 22, 818. [Google Scholar] [CrossRef] [PubMed]
  142. Petito-da-Silva, T.I.; Souza-Mello, V.; Barbosa-da-Silva, S. Empaglifozin Mitigates NAFLD in High-Fat-Fed Mice by Alleviating Insulin Resistance, Lipogenesis and ER Stress. Mol. Cell. Endocrinol. 2019, 498, 110539. [Google Scholar] [CrossRef] [PubMed]
  143. Shih, J.Y.; Lin, Y.W.; Fisch, S.; Cheng, J.T.; Kang, N.W.; Hong, C.S.; Chen, Z.C.; Chang, W.T. Dapagliflozin Suppresses ER Stress and Improves Subclinical Myocardial Function in Diabetes: From Bedside to Bench. Diabetes 2021, 70, 262–267. [Google Scholar] [CrossRef] [PubMed]
  144. Uthman, L.; Kuschma, M.; Römer, G.; Boomsma, M.; Kessler, J.; Hermanides, J.; Hollmann, M.W.; Preckel, B.; Zuurbier, C.J.; Weber, N.C. Novel Anti-Inflammatory Effects of Canagliflozin Involving Hexokinase II in Lipopolysaccharide-Stimulated Human Coronary Artery Endothelial Cells. Cardiovasc. Drugs Ther. 2021, 35, 1083–1094. [Google Scholar] [CrossRef]
  145. Abdollahi, E.; Keyhanfar, F.; Delbandi, A.-A.; Falak, R.; Hajimiresmaiel, S.J.; Shafiei, M. Dapagliflozin Exerts Anti-Inflammatory Effects via Inhibition of LPS-Induced TLR-4 Overexpression and NF-ΚB Activation in Human Endothelial Cells and Differentiated Macrophages. Eur. J. Pharmacol. 2022, 918, 174715. [Google Scholar] [CrossRef]
  146. Tian, Y.; Gao, L.; Guo, Y.; Xu, Y. Short-Term Phlorizin Treatment Attenuates Adipose Tissue Inflammation without Alerting Obesity in High-Fat Diet Fed Mice. J. Food Biochem. 2017, 41, e12407. [Google Scholar] [CrossRef]
  147. Kamdi, S.P.; Raval, A.; Nakhate, K.T. Phloridzin Attenuates Lipopolysaccharide-Induced Cognitive Impairment via Antioxidant, Anti-Inflammatory and Neuromodulatory Activities. Cytokine 2021, 139, 155408. [Google Scholar] [CrossRef]
  148. Chang, W.T.; Huang, W.C.; Liou, C.J. Evaluation of the Anti-Inflammatory Effects of Phloretin and Phlorizin in Lipopolysaccharide-Stimulated Mouse Macrophages. Food Chem. 2012, 134, 972–979. [Google Scholar] [CrossRef]
  149. Leyva-López, N.; Gutierrez-Grijalva, E.; Ambriz-Perez, D.; Heredia, J. Flavonoids as Cytokine Modulators: A Possible Therapy for Inflammation-Related Diseases. Int. J. Mol. Sci. 2016, 17, 921. [Google Scholar] [CrossRef]
  150. Jasim, H.A.; Nahar, L.; Jasim, M.A.; Moore, S.A.; Ritchie, K.J.; Sarker, S.D. Chalcones: Synthetic Chemistry Follows Where Nature Leads. Biomolecules 2021, 11, 1203. [Google Scholar] [CrossRef] [PubMed]
  151. Nile, S.H.; Keum, Y.S.; Nile, A.S.; Jalde, S.S.; Patel, R.V. Antioxidant, Anti-Inflammatory, and Enzyme Inhibitory Activity of Natural Plant Flavonoids and Their Synthesized Derivatives. J. Biochem. Mol. Toxicol. 2018, 32, e22002. [Google Scholar] [CrossRef] [PubMed]
  152. De Nardo, D.; Latz, E. NLRP3 Inflammasomes Link Inflammation and Metabolic Disease. Trends Immunol. 2011, 32, 373–379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Hotamisligil, G.S. Inflammation and Metabolic Disorders. Nature 2006, 444, 860–867. [Google Scholar] [CrossRef] [PubMed]
  154. Saltiel, A.R.; Olefsky, J.M. Inflammatory Mechanisms Linking Obesity and Metabolic Disease. J. Clin. Investig. 2017, 127, 1–4. [Google Scholar] [CrossRef]
  155. Wu, H.; Ballantyne, C.M. Metabolic Inflammation and Insulin Resistance in Obesity. Circ. Res. 2020, 126, 1549. [Google Scholar] [CrossRef]
  156. Blüher, M. Adipose Tissue Inflammation: A Cause or Consequence of Obesity-Related Insulin Resistance? Clin. Sci. 2016, 130, 1603–1614. [Google Scholar] [CrossRef]
  157. Mastrocola, R.; Aragno, M.; Alloatti, G.; Collino, M.; Penna, C.; Pagliaro, P. Metaflammation: Tissue-Specific Alterations of the NLRP3 Inflammasome Platform in Metabolic Syndrome. Curr. Med. Chem. 2017, 25, 1294–1310. [Google Scholar] [CrossRef]
  158. Hotamisligil, G.S. Inflammation, Metaflammation and Immunometabolic Disorders. Nature 2017, 542, 177–185. [Google Scholar] [CrossRef]
  159. Scheen, A.J. Sodium-Glucose Cotransporter Type 2 Inhibitors for the Treatment of Type 2 Diabetes Mellitus. Nat. Rev. Endocrinol. 2020, 16, 556–577. [Google Scholar] [CrossRef]
  160. Wang, X.; Ni, J.; Guo, R.; Li, L.; Su, J.; He, F.; Fan, G. SGLT2 Inhibitors Break the Vicious Circle between Heart Failure and Insulin Resistance: Targeting Energy Metabolism. Heart Fail. Rev. 2021, 27, 961–980. [Google Scholar] [CrossRef] [PubMed]
  161. Li, N.; Zhou, H. SGLT2 Inhibitors: A Novel Player in the Treatment and Prevention of Diabetic Cardiomyopathy. Drug Des. Devel. Ther. 2020, 14, 4775. [Google Scholar] [CrossRef] [PubMed]
  162. DeFronzo, R.A.; Reeves, W.B.; Awad, A.S. Pathophysiology of Diabetic Kidney Disease: Impact of SGLT2 Inhibitors. Nat. Rev. Nephrol. 2021, 17, 319–334. [Google Scholar] [CrossRef] [PubMed]
  163. Gharaibeh, N.E.; Rahhal, M.-N.; Rahimi, L.; Ismail-Beigi, F. SGLT-2 Inhibitors as Promising Therapeutics for Non-Alcoholic Fatty Liver Disease: Pathophysiology, Clinical Outcomes, and Future Directions. Diabetes Metab. Syndr. Obes. 2019, 12, 1001–1012. [Google Scholar] [CrossRef] [Green Version]
  164. Watanabe, S.; Alexander, M.; Misharin, A.V.; Budinger, G.R.S. The Role of Macrophages in the Resolution of Inflammation. J. Clin. Investig. 2019, 129, 2619–2628. [Google Scholar] [CrossRef] [Green Version]
  165. Shapouri-Moghaddam, A.; Mohammadian, S.; Vazini, H.; Taghadosi, M.; Esmaeili, S.A.; Mardani, F.; Seifi, B.; Mohammadi, A.; Afshari, J.T.; Sahebkar, A. Macrophage Plasticity, Polarization, and Function in Health and Disease. J. Cell. Physiol. 2018, 233, 6425–6440. [Google Scholar] [CrossRef] [PubMed]
  166. Kuzmich, N.; Sivak, K.; Chubarev, V.; Porozov, Y.; Savateeva-Lyubimova, T.; Peri, F. TLR4 Signaling Pathway Modulators as Potential Therapeutics in Inflammation and Sepsis. Vaccines 2017, 5, 34. [Google Scholar] [CrossRef] [Green Version]
  167. Liu, T.; Zhang, L.; Joo, D.; Sun, S.C. NF-ΚB Signaling in Inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef] [Green Version]
  168. Gordon, S.; Martinez, F.O. Alternative Activation of Macrophages: Mechanism and Functions. Immunity 2010, 32, 593–604. [Google Scholar] [CrossRef] [Green Version]
  169. Lee, T.-M.; Chang, N.-C.; Lin, S.-Z. Dapagliflozin, a Selective SGLT2 Inhibitor, Attenuated Cardiac Fibrosis by Regulating the Macrophage Polarization via STAT3 Signaling in Infarcted Rat Hearts. Free Radic. Biol. Med. 2017, 104, 298–310. [Google Scholar] [CrossRef]
  170. Leng, W.; Ouyang, X.; Lei, X.; Wu, M.; Chen, L.; Wu, Q.; Deng, W.; Liang, Z. The SGLT-2 Inhibitor Dapagliflozin Has a Therapeutic Effect on Atherosclerosis in Diabetic ApoE −/− Mice. Mediat. Inflamm. 2016, 2016, 6305735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Terasaki, M.; Hiromura, M.; Mori, Y.; Kohashi, K.; Nagashima, M.; Kushima, H.; Watanabe, T.; Hirano, T. Amelioration of Hyperglycemia with a Sodium-Glucose Cotransporter 2 Inhibitor Prevents Macrophage-Driven Atherosclerosis through Macrophage Foam Cell Formation Suppression in Type 1 and Type 2 Diabetic Mice. PLoS ONE 2015, 10, e0143396. [Google Scholar] [CrossRef] [PubMed]
  172. Gaspari, T.; Spizzo, I.; Liu, H.B.; Hu, Y.; Simpson, R.W.; Widdop, R.E.; Dear, A.E. Dapagliflozin Attenuates Human Vascular Endothelial Cell Activation and Induces Vasorelaxation: A Potential Mechanism for Inhibition of Atherogenesis. Diabetes Vasc. Dis. Res. 2018, 15, 64–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Lee, S.G.; Lee, S.J.; Lee, J.J.; Kim, J.S.; Lee, O.H.; Kim, C.K.; Kim, D.; Lee, Y.H.; Oh, J.; Park, S.; et al. Anti-Inflammatory Effect for Atherosclerosis Progression by Sodium-Glucose Cotransporter 2 (SGLT-2) Inhibitor in a Normoglycemic Rabbit Model. Korean Circ. J. 2020, 50, 443–457. [Google Scholar] [CrossRef]
  174. Kohlmorgen, C.; Gerfer, S.; Feldmann, K.; Twarock, S.; Hartwig, S.; Lehr, S.; Klier, M.; Krüger, I.; Helten, C.; Keul, P.; et al. Dapagliflozin Reduces Thrombin Generation and Platelet Activation: Implications for Cardiovascular Risk Reduction in Type 2 Diabetes Mellitus. Diabetologia 2021, 64, 1834–1849. [Google Scholar] [CrossRef]
  175. Stelmaszyk, A.; Wesołowska, A.; Pomieczyńska, K.; Iskakova, S.; Frydrychowicz, M.; Dworacki, G.; Dworacka, M. The Impact of Dapagliflozin on Glucose Excursions Related to Early Proatherogenic Derangement in the Aortic Wall. Saudi Pharm. J. 2018, 26, 1192–1198. [Google Scholar] [CrossRef]
  176. Liu, H.; Wei, P.; Fu, W.; Xia, C.; Li, Y.; Tian, K.; Li, Y.; Cheng, D.; Sun, J.; Xu, Y.; et al. Dapagliflozin Ameliorates the Formation and Progression of Experimental Abdominal Aortic Aneurysms by Reducing Aortic Inflammation in Mice. Oxid. Med. Cell. Longev. 2022, 2022, 8502059. [Google Scholar] [CrossRef]
  177. Leng, W.; Wu, M.; Pan, H.; Lei, X.; Chen, L.; Wu, Q.; Ouyang, X.; Liang, Z. The SGLT2 Inhibitor Dapagliflozin Attenuates the Activity of ROS-NLRP3 Inflammasome Axis in Steatohepatitis with Diabetes Mellitus. Ann. Transl. Med. 2019, 7, 429. [Google Scholar] [CrossRef]
  178. Tang, L.; Wu, Y.; Tian, M.; Sjöström, C.D.; Johansson, U.; Peng, X.R.; Smith, D.M.; Huang, Y. Dapagliflozin Slows the Progression of the Renal and Liver Fibrosis Associated with Type 2 Diabetes. Am. J. Physiol.-Endocrinol. Metab. 2017, 313, E563–E576. [Google Scholar] [CrossRef]
  179. Terami, N.; Ogawa, D.; Tachibana, H.; Hatanaka, T.; Wada, J.; Nakatsuka, A.; Eguchi, J.; Sato Horiguchi, C.; Nishii, N.; Yamada, H.; et al. Long-Term Treatment with the Sodium Glucose Cotransporter 2 Inhibitor, Dapagliflozin, Ameliorates Glucose Homeostasis and Diabetic Nephropathy in Db/Db Mice. PLoS ONE 2014, 9, e100777. [Google Scholar] [CrossRef]
  180. Hatanaka, T.; Ogawa, D.; Tachibana, H.; Eguchi, J.; Inoue, T.; Yamada, H.; Takei, K.; Makino, H.; Wada, J. Inhibition of SGLT2 Alleviates Diabetic Nephropathy by Suppressing High Glucose-Induced Oxidative Stress in Type 1 Diabetic Mice. Pharmacol. Res. Perspect. 2016, 4, e00239. [Google Scholar] [CrossRef] [PubMed]
  181. Shin, S.J.; Chung, S.; Kim, S.J.; Lee, E.M.; Yoo, Y.H.; Kim, J.W.; Ahn, Y.B.; Kim, E.S.; Moon, S.D.; Kim, M.J.; et al. Effect of Sodium-Glucose Co-Transporter 2 Inhibitor, Dapagliflozin, on Renal Renin-Angiotensin System in an Animal Model of Type 2 Diabetes. PLoS ONE 2016, 11, e0165703. [Google Scholar] [CrossRef] [PubMed]
  182. Cassis, P.; Locatelli, M.; Cerullo, D.; Corna, D.; Buelli, S.; Zanchi, C.; Villa, S.; Morigi, M.; Remuzzi, G.; Benigni, A.; et al. SGLT2 Inhibitor Dapagliflozin Limits Podocyte Damage in Proteinuric Nondiabetic Nephropathy. JCI Insight 2018, 3. [Google Scholar] [CrossRef] [Green Version]
  183. Lin, F.; Song, C.; Zeng, Y.; Li, Y.; Li, H.; Liu, B.; Dai, M.; Pan, P. Canagliflozin Alleviates LPS-Induced Acute Lung Injury by Modulating Alveolar Macrophage Polarization. Int. Immunopharmacol. 2020, 88, 106969. [Google Scholar] [CrossRef] [PubMed]
  184. Shiba, K.; Tsuchiya, K.; Komiya, C.; Miyachi, Y.; Mori, K.; Shimazu, N.; Yamaguchi, S.; Ogasawara, N.; Katoh, M.; Itoh, M.; et al. Canagliflozin, an SGLT2 Inhibitor, Attenuates the Development of Hepatocellular Carcinoma in a Mouse Model of Human NASH. Sci. Rep. 2018, 8, 2362. [Google Scholar] [CrossRef] [Green Version]
  185. Naznin, F.; Sakoda, H.; Okada, T.; Tsubouchi, H.; Waise, T.M.Z.; Arakawa, K.; Nakazato, M. Canagliflozin, a Sodium Glucose Cotransporter 2 Inhibitor, Attenuates Obesity-Induced Inflammation in the Nodose Ganglion, Hypothalamus, and Skeletal Muscle of Mice. Eur. J. Pharmacol. 2017, 794, 37–44. [Google Scholar] [CrossRef]
  186. Nasiri-Ansari, Ν.; Dimitriadis, G.K.; Agrogiannis, G.; Perrea, D.; Kostakis, I.D.; Kaltsas, G.; Papavassiliou, A.G.; Randeva, H.S.; Kassi, E. Canagliflozin Attenuates the Progression of Atherosclerosis and Inflammation Process in APOE Knockout Mice. Cardiovasc. Diabetol. 2018, 17, 106. [Google Scholar] [CrossRef] [Green Version]
  187. Woods, T.C.; Satou, R.; Miyata, K.; Katsurada, A.; Dugas, C.M.; Klingenberg, N.C.; Fonseca, V.A.; Navar, L.G. Canagliflozin Prevents Intrarenal Angiotensinogen Augmentation and Mitigates Kidney Injury and Hypertension in Mouse Model of Type 2 Diabetes Mellitus. Am. J. Nephrol. 2019, 49, 331–342. [Google Scholar] [CrossRef]
  188. Inoue, M.K.; Matsunaga, Y.; Nakatsu, Y.; Yamamotoya, T.; Ueda, K.; Kushiyama, A.; Sakoda, H.; Fujishiro, M.; Ono, H.; Iwashita, M.; et al. Possible Involvement of Normalized Pin1 Expression Level and AMPK Activation in the Molecular Mechanisms Underlying Renal Protective Effects of SGLT2 Inhibitors in Mice. Diabetol. Metab. Syndr. 2019, 11, 57. [Google Scholar] [CrossRef]
  189. Pennig, J.; Scherrer, P.; Gissler, M.C.; Anto-Michel, N.; Hoppe, N.; Füner, L.; Härdtner, C.; Stachon, P.; Wolf, D.; Hilgendorf, I.; et al. Glucose Lowering by SGLT2-Inhibitor Empagliflozin Accelerates Atherosclerosis Regression in Hyperglycemic STZ-Diabetic Mice. Sci. Rep. 2019, 9, 17937. [Google Scholar] [CrossRef]
  190. Lee, N.; Heo, Y.J.; Choi, S.-E.E.; Jeon, J.Y.; Han, S.J.; Kim, D.J.; Kang, Y.; Lee, K.W.; Kim, H.J. Anti-Inflammatory Effects of Empagliflozin and Gemigliptin on LPS-Stimulated Macrophage via the IKK/NF-ΚB, MKK7/JNK, and JAK2/STAT1 Signalling Pathways. J. Immunol. Res. 2021, 2021, 9944880. [Google Scholar] [CrossRef]
  191. Koyani, C.N.; Plastira, I.; Sourij, H.; Hallström, S.; Schmidt, A.; Rainer, P.P.; Bugger, H.; Frank, S.; Malle, E.; von Lewinski, D. Empagliflozin Protects Heart from Inflammation and Energy Depletion via AMPK Activation. Pharmacol. Res. 2020, 158, 104870. [Google Scholar] [CrossRef] [PubMed]
  192. Ganbaatar, B.; Fukuda, D.; Shinohara, M.; Yagi, S.; Kusunose, K.; Yamada, H.; Soeki, T.; Hirata, K.-I.; Sata, M. Empagliflozin Ameliorates Endothelial Dysfunction and Suppresses Atherogenesis in Diabetic Apolipoprotein E-Deficient Mice. Eur. J. Pharmacol. 2020, 875, 173040. [Google Scholar] [CrossRef]
  193. Ortega, R.; Collado, A.; Selles, F.; Gonzalez-Navarro, H.; Sanz, M.J.; Real, J.T.; Piqueras, L. SGLT-2 (Sodium-Glucose Cotransporter 2) Inhibition Reduces Ang II (Angiotensin II)-Induced Dissecting Abdominal Aortic Aneurysm in ApoE (Apolipoprotein E) Knockout Mice. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 1614–1628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Radlinger, B.; Hornsteiner, F.; Folie, S.; Salvenmoser, W.; Haubner, B.J.; Schuetz, T.; Haas, S.; Ress, C.; Adolph, T.E.; Salzmann, K.; et al. Cardioprotective Effects of Short-Term Empagliflozin Treatment in Db/Db Mice. Sci. Rep. 2020, 10, 19686. [Google Scholar] [CrossRef] [PubMed]
  195. Lin, B.; Koibuchi, N.; Hasegawa, Y.; Sueta, D.; Toyama, K.; Uekawa, K.; Ma, M.; Nakagawa, T.; Kusaka, H.; Kim-Mitsuyama, S. Glycemic Control with Empagliflozin, a Novel Selective SGLT2 Inhibitor, Ameliorates Cardiovascular Injury and Cognitive Dysfunction in Obese and Type 2 Diabetic Mice. Cardiovasc. Diabetol. 2014, 13, 148. [Google Scholar] [CrossRef] [Green Version]
  196. Byrne, N.J.; Matsumura, N.; Maayah, Z.H.; Ferdaoussi, M.; Takahara, S.; Darwesh, A.M.; Levasseur, J.L.; Jahng, J.W.S.; Vos, D.; Parajuli, N.; et al. Empagliflozin Blunts Worsening Cardiac Dysfunction Associated With Reduced NLRP3 (Nucleotide-Binding Domain-Like Receptor Protein 3) Inflammasome Activation in Heart Failure. Circ. Heart Fail. 2020, 13, e006277. [Google Scholar] [CrossRef]
  197. Kräker, K.; Herse, F.; Golic, M.; Reichhart, N.; Crespo-Garcia, S.; Strauß, O.; Grune, J.; Kintscher, U.; Ebrahim, M.; Bader, M.; et al. Effects of Empagliflozin and Target-Organ Damage in a Novel Rodent Model of Heart Failure Induced by Combined Hypertension and Diabetes. Sci. Rep. 2020, 10, 14061. [Google Scholar] [CrossRef]
  198. Zhang, Q.-Q.; Li, G.-Q.; Zhong, Y.; Wang, J.; Wang, A.-N.; Zhou, X.; Mao, X.-M. Empagliflozin Improves Chronic Hypercortisolism-Induced Abnormal Myocardial Structure and Cardiac Function in Mice. Ther. Adv. Chronic Dis. 2020, 11, 2040622320974833. [Google Scholar] [CrossRef]
  199. Meng, Z.; Liu, X.; Li, T.; Fang, T.; Cheng, Y.; Han, L.; Sun, B.; Chen, L. The SGLT2 Inhibitor Empagliflozin Negatively Regulates IL-17/IL-23 Axis-Mediated Inflammatory Responses in T2DM with NAFLD via the AMPK/MTOR/Autophagy Pathway. Int. Immunopharmacol. 2021, 94, 107492. [Google Scholar] [CrossRef]
  200. Lee, N.; Heo, Y.J.; Choi, S.E.; Jeon, J.Y.; Han, S.J.; Kim, D.J.; Kang, Y.; Lee, K.W.; Kim, H.J. Hepatoprotective Effects of Gemigliptin and Empagliflozin in a Murine Model of Diet-Induced Non-Alcoholic Fatty Liver Disease. Biochem. Biophys. Res. Commun. 2022, 588, 154–160. [Google Scholar] [CrossRef] [PubMed]
  201. Jojima, T.; Tomotsune, T.; Iijima, T.; Akimoto, K.; Suzuki, K.; Aso, Y. Empagliflozin (an SGLT2 Inhibitor), Alone or in Combination with Linagliptin (a DPP-4 Inhibitor), Prevents Steatohepatitis in a Novel Mouse Model of Non-Alcoholic Steatohepatitis and Diabetes. Diabetol. Metab. Syndr. 2016, 8, 45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Ojima, A.; Matsui, T.; Nishino, Y.; Nakamura, N.; Yamagishi, S. Empagliflozin, an Inhibitor of Sodium-Glucose Cotransporter 2 Exerts Anti-Inflammatory and Antifibrotic Effects on Experimental Diabetic Nephropathy Partly by Suppressing AGEs-Receptor Axis. Horm. Metab. Res. 2015, 47, 686–692. [Google Scholar] [CrossRef] [Green Version]
  203. Hudkins, K.L.; Li, X.; Holland, A.L.; Swaminathan, S.; Alpers, C.E. Regression of Diabetic Nephropathy by Treatment with Empagliflozin in BTBR Ob/Ob Mice. Nephrol. Dial. Transplant. 2022, 37, 847–859. [Google Scholar] [CrossRef]
  204. Maayah, Z.H.; Ferdaoussi, M.; Takahara, S.; Soni, S.; Dyck, J.R.B. Empagliflozin Suppresses Inflammation and Protects against Acute Septic Renal Injury. Inflammopharmacology 2021, 29, 269–279. [Google Scholar] [CrossRef]
  205. Castoldi, G.; Carletti, R.; Ippolito, S.; Colzani, M.; Barzaghi, F.; Stella, A.; Zerbini, G.; Perseghin, G.; Zatti, G.; di Gioia, C.R.T. Sodium-Glucose Cotransporter 2 Inhibition Prevents Renal Fibrosis in Cyclosporine Nephropathy. Acta Diabetol. 2021, 58, 1059–1070. [Google Scholar] [CrossRef] [PubMed]
  206. Han, J.H.; Oh, T.J.; Lee, G.; Maeng, H.J.; Lee, D.H.; Kim, K.M.; Choi, S.H.; Jang, H.C.; Lee, H.S.; Park, K.S.; et al. The Beneficial Effects of Empagliflozin, an SGLT2 Inhibitor, on Atherosclerosis in ApoE −/− Mice Fed a Western Diet. Diabetologia 2017, 60, 364–376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Xu, L.; Nagata, N.; Nagashimada, M.; Zhuge, F.; Ni, Y.; Chen, G.; Mayoux, E.; Kaneko, S.; Ota, T. SGLT2 Inhibition by Empagliflozin Promotes Fat Utilization and Browning and Attenuates Inflammation and Insulin Resistance by Polarizing M2 Macrophages in Diet-Induced Obese Mice. EBioMedicine 2017, 20, 137–149. [Google Scholar] [CrossRef] [Green Version]
  208. Xu, L.; Nagata, N.; Chen, G.; Nagashimada, M.; Zhuge, F.; Ni, Y.; Sakai, Y.; Kaneko, S.; Ota, T. Empagliflozin Reverses Obesity and Insulin Resistance through Fat Browning and Alternative Macrophage Activation in Mice Fed a High-Fat Diet. BMJ Open Diabetes Res. Care 2019, 7, e000783. [Google Scholar] [CrossRef]
  209. Nakatsu, Y.; Kokubo, H.; Bumdelger, B.; Yoshizumi, M.; Yamamotoya, T.; Matsunaga, Y.; Ueda, K.; Inoue, Y.; Inoue, M.-K.; Fujishiro, M.; et al. The SGLT2 Inhibitor Luseogliflozin Rapidly Normalizes Aortic MRNA Levels of Inflammation-Related but Not Lipid-Metabolism-Related Genes and Suppresses Atherosclerosis in Diabetic ApoE KO Mice. Int. J. Mol. Sci. 2017, 18, 1704. [Google Scholar] [CrossRef] [Green Version]
  210. Mori, K.; Tsuchiya, K.; Nakamura, S.; Miyachi, Y.; Shiba, K.; Ogawa, Y.; Kitamura, K. Ipragliflozin-Induced Adipose Expansion Inhibits Cuff-Induced Vascular Remodeling in Mice. Cardiovasc. Diabetol. 2019, 18, 83. [Google Scholar] [CrossRef] [PubMed]
  211. Salim, H.M.; Fukuda, D.; Yagi, S.; Soeki, T.; Shimabukuro, M.; Sata, M. Glycemic Control with Ipragliflozin, a Novel Selective SGLT2 Inhibitor, Ameliorated Endothelial Dysfunction in Streptozotocin-Induced Diabetic Mouse. Front. Cardiovasc. Med. 2016, 3, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Mori, Y.; Terasaki, M.; Hiromura, M.; Saito, T.; Kushima, H.; Koshibu, M.; Osaka, N.; Ohara, M.; Fukui, T.; Ohtaki, H.; et al. Luseogliflozin Attenuates Neointimal Hyperplasia after Wire Injury in High-Fat Diet-Fed Mice via Inhibition of Perivascular Adipose Tissue Remodeling. Cardiovasc. Diabetol. 2019, 18, 143. [Google Scholar] [CrossRef] [PubMed]
  213. Miyachi, Y.; Tsuchiya, K.; Shiba, K.; Mori, K.; Komiya, C.; Ogasawara, N.; Ogawa, Y. A Reduced M1-like/M2-like Ratio of Macrophages in Healthy Adipose Tissue Expansion during SGLT2 Inhibition. Sci. Rep. 2018, 8, 16113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Komiya, C.; Tsuchiya, K.; Shiba, K.; Miyachi, Y.; Furuke, S.; Shimazu, N.; Yamaguchi, S.; Kanno, K.; Ogawa, Y. Ipragliflozin Improves Hepatic Steatosis in Obese Mice and Liver Dysfunction in Type 2 Diabetic Patients Irrespective of Body Weight Reduction. PLoS ONE 2016, 11, e0151511. [Google Scholar] [CrossRef] [Green Version]
  215. Okuma, H.; Mori, K.; Nakamura, S.; Sekine, T.; Ogawa, Y.; Tsuchiya, K. Ipragliflozin Ameliorates Diabetic Nephropathy Associated with Perirenal Adipose Expansion in Mice. Int. J. Mol. Sci. 2021, 22, 7329. [Google Scholar] [CrossRef]
  216. The Human Protein Atlas Single Cell Type—NLRP3. Available online: https://www.proteinatlas.org/ENSG00000162711-NLRP3/single+cell+type (accessed on 28 April 2022).
  217. Marcuzzi, A.; Melloni, E.; Zauli, G.; Romani, A.; Secchiero, P.; Maximova, N.; Rimondi, E. Autoinflammatory Diseases and Cytokine Storms-Imbalances of Innate and Adaptative Immunity. Int. J. Mol. Sci. 2021, 22, 11241. [Google Scholar] [CrossRef]
  218. Swanson, K.V.; Deng, M.; Ting, J.P.Y. The NLRP3 Inflammasome: Molecular Activation and Regulation to Therapeutics. Nat. Rev. Immunol. 2019, 19, 477–489. [Google Scholar] [CrossRef]
  219. Burdette, B.E.; Esparza, A.N.; Zhu, H.; Wang, S. Gasdermin D in Pyroptosis. Acta Pharm. Sin. B 2021, 11, 2768–2782. [Google Scholar] [CrossRef]
  220. Lee, H.M.; Kim, J.J.; Kim, H.J.; Shong, M.; Ku, B.J.; Jo, E.K. Upregulated NLRP3 Inflammasome Activation in Patients With Type 2 Diabetes. Diabetes 2013, 62, 194–204. [Google Scholar] [CrossRef] [Green Version]
  221. Abbate, A.; Toldo, S.; Marchetti, C.; Kron, J.; Van Tassell, B.W.; Dinarello, C.A. Interleukin-1 and the Inflammasome as Therapeutic Targets in Cardiovascular Disease. Circ. Res. 2020, 126, 1260–1280. [Google Scholar] [CrossRef] [PubMed]
  222. Xiong, W.; Meng, X.-F.; Zhang, C. NLRP3 Inflammasome in Metabolic-Associated Kidney Diseases: An Update. Front. Immunol. 2021, 12, 714340. [Google Scholar] [CrossRef] [PubMed]
  223. Al Mamun, A.; Akter, A.; Hossain, S.; Sarker, T.; Safa, S.A.; Mustafa, Q.G.; Muhammad, S.A.; Munir, F. Role of NLRP3 Inflammasome in Liver Disease. J. Dig. Dis. 2020, 21, 430–436. [Google Scholar] [CrossRef] [PubMed]
  224. Ye, Y.; Bajaj, M.; Yang, H.C.; Perez-Polo, J.R.; Birnbaum, Y. SGLT-2 Inhibition with Dapagliflozin Reduces the Activation of the Nlrp3/ASC Inflammasome and Attenuates the Development of Diabetic Cardiomyopathy in Mice with Type 2 Diabetes. Further Augmentation of the Effects with Saxagliptin, a DPP4 Inhibitor. Cardiovasc. Drugs Ther. 2017, 31, 119–132. [Google Scholar] [CrossRef] [PubMed]
  225. El-Rous, M.A.; Saber, S.; Raafat, E.M.; Ahmed, A.A.E. Dapagliflozin, an SGLT2 Inhibitor, Ameliorates Acetic Acid-Induced Colitis in Rats by Targeting NFκB/AMPK/NLRP3 Axis. Inflammopharmacology 2021, 29, 1169–1185. [Google Scholar] [CrossRef]
  226. Muhammad, R.N.; Ahmed, L.A.; Abdul Salam, R.M.; Ahmed, K.A.; Attia, A.S. Crosstalk Among NLRP3 Inflammasome, ET B R Signaling, and MiRNAs in Stress-Induced Depression-Like Behavior: A Modulatory Role for SGLT2 Inhibitors. Neurotherapeutics 2021, 18, 2664–2681. [Google Scholar] [CrossRef]
  227. Ke, Q.; Shi, C.; Lv, Y.; Wang, L.; Luo, J.; Jiang, L.; Yang, J.; Zhou, Y. SGLT2 Inhibitor Counteracts NLRP3 Inflammasome via Tubular Metabolite Itaconate in Fibrosis Kidney. FASEB J. 2022, 36, e22078. [Google Scholar] [CrossRef]
  228. Birnbaum, Y.; Bajaj, M.; Yang, H.C.; Ye, Y. Combined SGLT2 and DPP4 Inhibition Reduces the Activation of the Nlrp3/ASC Inflammasome and Attenuates the Development of Diabetic Nephropathy in Mice with Type 2 Diabetes. Cardiovasc. Drugs Ther. 2018, 32, 135–145. [Google Scholar] [CrossRef]
  229. Niu, Y.; Zhang, Y.; Zhang, W.; Lu, J.; Chen, Y.; Hao, W.; Zhou, J.; Wang, L.; Xie, W. Canagliflozin Ameliorates NLRP3 Inflammasome-Mediated Inflammation through Inhibiting NF-ΚB Signaling and Upregulating Bif-1. Front. Pharmacol. 2022, 13, 820541. [Google Scholar] [CrossRef]
  230. Kim, S.R.; Lee, S.-G.; Kim, S.H.; Kim, J.H.; Choi, E.; Cho, W.; Rim, J.H.; Hwang, I.; Lee, C.J.; Lee, M.; et al. SGLT2 Inhibition Modulates NLRP3 Inflammasome Activity via Ketones and Insulin in Diabetes with Cardiovascular Disease. Nat. Commun. 2020, 11, 2127. [Google Scholar] [CrossRef]
  231. Sukhanov, S.; Higashi, Y.; Yoshida, T.; Mummidi, S.; Aroor, A.R.; Jeffrey Russell, J.; Bender, S.B.; DeMarco, V.G.; Chandrasekar, B. The SGLT2 Inhibitor Empagliflozin Attenuates Interleukin-17A-Induced Human Aortic Smooth Muscle Cell Proliferation and Migration by Targeting TRAF3IP2/ROS/NLRP3/Caspase-1-Dependent IL-1β and IL-18 Secretion. Cell. Signal. 2021, 77, 109825. [Google Scholar] [CrossRef] [PubMed]
  232. Deguine, J.; Barton, G.M. MyD88: A Central Player in Innate Immune Signaling. F1000Prime Rep. 2014, 6, 97. [Google Scholar] [CrossRef]
  233. Quagliariello, V.; De Laurentiis, M.; Rea, D.; Barbieri, A.; Monti, M.G.; Carbone, A.; Paccone, A.; Altucci, L.; Conte, M.; Canale, M.L.; et al. The SGLT-2 Inhibitor Empagliflozin Improves Myocardial Strain, Reduces Cardiac Fibrosis and pro-Inflammatory Cytokines in Non-Diabetic Mice Treated with Doxorubicin. Cardiovasc. Diabetol. 2021, 20, 150. [Google Scholar] [CrossRef] [PubMed]
  234. Benetti, E.; Mastrocola, R.; Vitarelli, G.; Cutrin, J.C.; Nigro, D.; Chiazza, F.; Mayoux, E.; Collino, M.; Fantozzi, R. Empagliflozin Protects against Diet-Induced NLRP-3 Inflammasome Activation and Lipid Accumulation. J. Pharmacol. Exp. Ther. 2016, 359, 45–53. [Google Scholar] [CrossRef] [PubMed]
  235. Liu, P.; Zhang, Z.; Wang, J.; Zhang, X.; Yu, X.; Li, Y. Empagliflozin Protects Diabetic Pancreatic Tissue from Damage by Inhibiting the Activation of the NLRP3/Caspase-1/GSDMD Pathway in Pancreatic β Cells: In Vitro and in Vivo Studies. Bioengineered 2021, 12, 9356–9366. [Google Scholar] [CrossRef]
  236. Xue, M.; Li, T.; Wang, Y.; Chang, Y.; Cheng, Y.; Lu, Y.; Liu, X.; Xu, L.; Li, X.; Yu, X.; et al. Empagliflozin Prevents Cardiomyopathy via SGC-CGMP-PKG Pathway in Type 2 Diabetes Mice. Clin. Sci. 2019, 133, 1705–1720. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Main sites of expression and function of SGLT1 and SGLT2. Glu: glucose. GLUT2: Facilitated Glucose Transporter Member 2. The figure was drawn using pictures from Servier Medical Art, which are licensed under a Creative Commons Attribution 3.0 Unported License (http://smart.servier.com/, 15 April 2022).
Figure 1. Main sites of expression and function of SGLT1 and SGLT2. Glu: glucose. GLUT2: Facilitated Glucose Transporter Member 2. The figure was drawn using pictures from Servier Medical Art, which are licensed under a Creative Commons Attribution 3.0 Unported License (http://smart.servier.com/, 15 April 2022).
Ijms 23 05634 g001
Figure 2. Main therapeutic targets of SGLT2i. The figure was drawn using pictures from Servier Medical Art, which are licensed under a Creative Commons Attribution 3.0 Unported License (http://smart.servier.com/, 21 April 2022).
Figure 2. Main therapeutic targets of SGLT2i. The figure was drawn using pictures from Servier Medical Art, which are licensed under a Creative Commons Attribution 3.0 Unported License (http://smart.servier.com/, 21 April 2022).
Ijms 23 05634 g002
Figure 3. Main anti-inflammatory effects of SGLT2i. The figure was drawn using pictures from Servier Medical Art, which are licensed under a Creative Commons Attribution 3.0 Unported License (http://smart.servier.com/, 21 April 2022).
Figure 3. Main anti-inflammatory effects of SGLT2i. The figure was drawn using pictures from Servier Medical Art, which are licensed under a Creative Commons Attribution 3.0 Unported License (http://smart.servier.com/, 21 April 2022).
Ijms 23 05634 g003
Table 2. Comparison between SGLT1 and SGLT2 co-transporters [42,43].
Table 2. Comparison between SGLT1 and SGLT2 co-transporters [42,43].
SGLT1SGLT2
Glucose affinityHigh (Km~0.5–2 mM)Low (Km~2–5 mM)
Glucose transport capacityLow (2 nmol/mg·min)High (10 nmol/mg·min)
Renal expressionS3 segmentS1 and S2 segments
Renal glucose reabsorption3–10%90–97%
Na+/glucose stoichiometry2:11:1
Expression in the small intestineYesNo
Main functionIntestinal absorption of glucose and galactose (urea and water)Renal reabsorption of glucose
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Feijóo-Bandín, S.; Aragón-Herrera, A.; Otero-Santiago, M.; Anido-Varela, L.; Moraña-Fernández, S.; Tarazón, E.; Roselló-Lletí, E.; Portolés, M.; Gualillo, O.; González-Juanatey, J.R.; et al. Role of Sodium-Glucose Co-Transporter 2 Inhibitors in the Regulation of Inflammatory Processes in Animal Models. Int. J. Mol. Sci. 2022, 23, 5634. https://doi.org/10.3390/ijms23105634

AMA Style

Feijóo-Bandín S, Aragón-Herrera A, Otero-Santiago M, Anido-Varela L, Moraña-Fernández S, Tarazón E, Roselló-Lletí E, Portolés M, Gualillo O, González-Juanatey JR, et al. Role of Sodium-Glucose Co-Transporter 2 Inhibitors in the Regulation of Inflammatory Processes in Animal Models. International Journal of Molecular Sciences. 2022; 23(10):5634. https://doi.org/10.3390/ijms23105634

Chicago/Turabian Style

Feijóo-Bandín, Sandra, Alana Aragón-Herrera, Manuel Otero-Santiago, Laura Anido-Varela, Sandra Moraña-Fernández, Estefanía Tarazón, Esther Roselló-Lletí, Manuel Portolés, Oreste Gualillo, José Ramón González-Juanatey, and et al. 2022. "Role of Sodium-Glucose Co-Transporter 2 Inhibitors in the Regulation of Inflammatory Processes in Animal Models" International Journal of Molecular Sciences 23, no. 10: 5634. https://doi.org/10.3390/ijms23105634

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop