ArticlePDF AvailableLiterature Review

The complex combination of COVID-19 and diabetes: pleiotropic changes in glucose metabolism

Authors:

Abstract and Figures

Purpose Angiotensin converting enzyme 2 (ACE2) is the door for SARS-CoV-2, expressed in critical metabolic tissues. So, it is rational that the new virus causes pleiotropic alterations in glucose metabolism, resulting in the complication of pre-existing diabetes’s pathophysiology or creating new disease mechanisms. However, it seems that less attention has been paid to this issue. This review aimed to highlight the importance of long-term consequences and pleiotropic alterations in glucose metabolism following COVID-19 and emphasize the need for basic and clinical research in metabolism and endocrinology. Results SARS-CoV-2 shifts cellular metabolism from oxidative phosphorylation to glycolysis, which leads to a decrease in ATP generation. Together with metabolic imbalance, the impaired immune system elevates the susceptibility of patients with diabetes to this deadly virus. SARS-CoV-2-induced metabolic alterations in immune cells can result in hyper inflammation and a cytokine storm. Metabolic dysfunction may affect therapies against SARS-CoV-2 infection. The effective control of metabolic complications could prove useful therapeutic targets for combating COVID-19. It is also necessary to understand the long-term consequences that will affect patients with diabetes who survived COVID-19. Conclusions Since the pathophysiology of COVID-19 is still mostly unknown, identifying the metabolic mechanisms contributing to its progression is essential to provide specific ways to prevent and improve this dangerous virus’s detrimental effects. The findings show that the new virus may induce new-onset diabetes with uncertain metabolic and clinical features, supporting a potential role of COVID-19 in the development of diabetes.
This content is subject to copyright. Terms and conditions apply.
Endocrine
https://doi.org/10.1007/s12020-021-02729-7
REVIEW
The complex combination of COVID-19 and diabetes: pleiotropic
changes in glucose metabolism
Abdolkarim Mahrooz 1,2 Giovanna Muscogiuri3Raffaella Buzzetti4Ernesto Maddaloni4
Received: 18 January 2021 / Accepted: 9 April 2021
© The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature 2021
Abstract
Purpose Angiotensin converting enzyme 2 (ACE2) is the door for SARS-CoV-2, expressed in critical metabolic tissues.
So, it is rational that the new virus causes pleiotropic alterations in glucose metabolism, resulting in the complication of pre-
existing diabetess pathophysiology or creating new disease mechanisms. However, it seems that less attention has been paid
to this issue. This review aimed to highlight the importance of long-term consequences and pleiotropic alterations in glucose
metabolism following COVID-19 and emphasize the need for basic and clinical research in metabolism and endocrinology.
Results SARS-CoV-2 shifts cellular metabolism from oxidative phosphorylation to glycolysis, which leads to a decrease in
ATP generation. Together with metabolic imbalance, the impaired immune system elevates the susceptibility of patients with
diabetes to this deadly virus. SARS-CoV-2-induced metabolic alterations in immune cells can result in hyper inammation
and a cytokine storm. Metabolic dysfunction may affect therapies against SARS-CoV-2 infection. The effective control of
metabolic complications could prove useful therapeutic targets for combating COVID-19. It is also necessary to understand
the long-term consequences that will affect patients with diabetes who survived COVID-19.
Conclusions Since the pathophysiology of COVID-19 is still mostly unknown, identifying the metabolic mechanisms
contributing to its progression is essential to provide specic ways to prevent and improve this dangerous viruss detrimental
effects. The ndings show that the new virus may induce new-onset diabetes with uncertain metabolic and clinical features,
supporting a potential role of COVID-19 in the development of diabetes.
Keywords COVID-19 SARS-CoV-2 Diabetes Glucose metabolism Long-term consequences
Introduction
The 2019 novel coronavirus disease (COVID-19) pan-
demic, caused by severe acute respiratory syndrome cor-
onavirus 2 (SARS-CoV-2), is seriously threatening health
systems. As a global healthcare issue, this pandemic has
posed a tremendous threat to humans and accounts for
causing considerable morbidity and mortality worldwide.
Diabetes is one of the most frequent comorbidity associated
with COVID-19 [1], and the highest mortality rates have
been registered in people with cardiometabolic disorders
[2]. Reports from the Centers for Disease Control and
Prevention (CDC) and other national health centers
demonstrated that the risk of a fatal outcome from COVID-
19 could be up to 50% higher in diabetes patients than
people without diabetes [3]. A meta-analysis conducted on
six studies with 1527 patients showed that the incidence of
diabetes was twofold greater in severe patients compared to
their non-severe counterparts [3]. An elevated risk of severe
complications, including Adult Respiratory Distress Syn-
drome and multiorgan failure, have been observed in
patients with diabetes [4,5]. Also, COVID-19 patients with
diabetes had an increased risk of mortality based on a recent
meta-analysis (OR =2.21, 95% CI: 1.832.66, p< 0.001)
[6]. Furthermore, the role of systemic endothelial dysfunc-
tion in the pathophysiology of COVID-19 in patients with
cardiometabolic disorders may be essential so that it may
*Abdolkarim Mahrooz
amahrooz@mazums.ac.ir
1Molecular and Cell Biology Research Center, Mazandaran
University of Medical Sciences, Sari, Iran
2Diabetes Research Center, Imam Khomeini Hospital, Mazandaran
University of Medical Sciences, Sari, Iran
3Sezione di Endocrinologia, Dipartimento di Medicina Clinica e
Chirurgia, Università Federico II Napoli, Napoli, Italy
4Department of Experimental Medicine, Sapienza University of
Rome, Rome, Italy
1234567890();,:
1234567890();,:
potentially be considered a new therapeutic target at mini-
mizing the severity of this infection [7].
SARS-CoV-2 infection is accompanied by the release of
many pro-inammatory cytokines [tumor necrosis factor
(TNF), interleukin-6 (IL-6), and IL-1β] described as cyto-
kine storm, which correlates with vascular hyperperme-
ability, lung injury, multiorgan failure, and COVID-19
severity [8,9]. Increased inammation and the massive
production of cytokines can generate insulin resistance and
also inuence beta-cell function, involving a further reduc-
tion of insulin secretion [10]. This may induce hyperglyce-
mia, which in turn can elevate the virulence of SARS-CoV-2
and reduces the phagocytic activity and polymorphonuclear
leukocytes [4,11]. Based on the ndings of Zhu et al. [12],
improved glycemic control had associated with better out-
comes in COVID-19 patients and pre-existing T2D, so that
virus people with well-controlled blood glucose (upper
limit 10 mmol/L) had markedly lower mortality in com-
parison to those with poorly controlled blood glucose (upper
limit > 10 mmol/L) during hospitalization.
On the other hand, it has also been suggested that
COVID-19 may trigger diabetes onset in predisposed sub-
jects. In this narrative review, we aim to describe this other
side of the coin, discussing the possible effects of COVID-19
on glucose homeostasis and related mechanisms.
Possible effects of COVID-19 on diabetes
SARS-CoV-2 infection in individuals with diabetes may
trigger stress and increase the release of hyperglycemic
hormones, such as catecholamines and glucocorticoids,
which would lead to increased blood glucose concentra-
tions and abnormal glucose variability [13], causing an
increased risk of metabolic emergencies. In particular,
COVID-19 can elevate the risk of both diabetic ketoaci-
dosis (DKA) and hyperosmolar hyperglycaemic state even
in people without previously recognized diabetes [14,15].
The risk of developing DKA is higher in people with T1D,
and additional metabolic complications can occur in these
patients [11,15]. The importance of ketoacidosis and
DKA can be prominent in children and adolescents. There
is a signicant increase in DKA and severe ketoacidosis at
diabetes diagnosis in children and adolescents during the
COVID-19 pandemic. More importantly, reduced medical
services, fear of approaching the health care system, and
more complex psychosocial factors may result in the
delayed DKA diagnosis, which can be life-threatening
[16]. Furthermore, more individuals with pre-existing type
2 diabetes (T2D) are progressing to insulin therapy with
severe COVID-19 [14]. Maddaloni et al. rst also hypo-
thesized that in the long term, the infection of pancreatic
beta-cells could trigger betacell autoimmunity [17], which
might trigger type 1 diabetes (T1D) onset in predisposed
subjects [18]. COVID-19 was associated with new and
persistent hyperglycemia so that some individuals with
normal HbA1C levels may develop new-onset diabetes
[19]. However, the extent and phenotype of new-onset
diabetes in patients with conrmed COVID-19 with a
previous negative history of diabetes remains to be
elucidated [11].
Some medications with a proven benecial effect in
COVID-19 and currently used for treating mild and severe
COVID-19 cases, such as corticosteroids, can exacerbate
glycemic control in patients with diabetes [11]. Although
hyperglycemia is usually considered the primary concern in
diabetes patients with COVID-19, hypoglycemic episodes
resulting from drug use, viral pathogenesis, and metabolic
perturbations of diabetes should not be ignored, particularly in
older patients and in those prone to hypoglycemia [20,21].
In addition to impaired glucose metabolism, diabetes is a
chronic inammatory disorder characterized by several
vascular and metabolic abnormalities [22]. It is expected
that SARS-CoV-2 infection could further worsen inam-
mation in individuals with diabetes. Together with meta-
bolic imbalance, the impaired immune system elevates the
susceptibility of patients with diabetes to pathogens such as
SARS-CoV-2 [23]. Several factors involved in the immune
dysfunction in diabetes patients, including hyperglycemia,
impaired T cell function, impaired macrophage function,
decreased neutrophil chemotaxis, and increased adherence
of microorganisms to diabetic cells [2327].
In T2D patients, an imbalance between coagulation and
brinolysis carries out with increased coagulant potential,
chronic platelet activation, and relative inhibition of
brinolysis, predisposing the patients to the development
of a hypercoagulable prothrombotic state [28]. This con-
dition can be exacerbated in patients with COVID-19.
Diabetes people with COVID-19 show a signicantly
higher D-dimer, a coagulation index, than those without
diabetes. Notably, the risk of coagulopathy and thrombosis
could be higher in diabetes and obesity, increasing sus-
ceptibility to thrombotic disorders [29]. Furthermore,
based on some evidence, endothelial injury and coagulo-
pathy may be central mediators of lung injury in SARS-
CoV-2 infection so that therapies that work on this basis
may be considered for COVID-19-related ARDS [30].
Gestational diabetes mellitus is one of the most common
medical problems found in pregnancy. Pregnant women
with diabetes may be more vulnerable to the severe effects
of SARS-CoV-2 infection [31,32]. It seems that there
are more clinical complications in pregnant women with
COVID-19, and new infection was associated with
enhanced preterm birth, preeclampsia, cesarean, and peri-
natal death [33]. Based on a case report study, pregnancy is
a high-risk period for euglycemic ketoacidosis, even in non-
diabetic women, particularly in infectious diseases such as
Endocrine
COVID-19 [34]. Drug-induced hyperglycemia or secondary
diabetes during COVID-19 treatment, particularly with
frequent use of corticosteroids, maybe one of the causes of
new-onset hyperglycemia in patients with SARS-CoV-2
infection. This can be observed especially in admitted
patients with COVID-19 without a history of dysglycemia
or diabetes and not on corticosteroids. New-onset diabetes
in COVID-19 patients has been related to signicantly
higher complications and all-cause death than subjects with
normoglycemia and preexisting diabetes [35].
The long-term consequences of SARS-CoV-2
infection should be more attentioned, particularly in
individuals with diabetes
Although the long-term consequences for patients recover-
ing from COVID-19 are unknown, researchers and health-
care professionals need to be aware of the importance of
continued monitoring of long-lasting consequences in
managing this strange disease. In this context, the results
obtained for previous coronaviruses may be useful. Long-
term outcomes following severe illness caused by SARS
and MERS were analyzed in a meta-analysis of 28 studies.
Accordingly, the increased prevalence of posttraumatic
stress disorder, depression, and anxiety were reported up to
6 months after discharge [36]. The ndings of a follow-up
study for 12 years on 25 recovered SARS patients also
showed that they tended to have increased hyperlipidemia,
cardiovascular abnormality, and abnormal glucose meta-
bolism, and signicant differences found in serum meta-
bolomes in comparison to healthy volunteers [37]. COVID-
19 survivors may, therefore, also experience long-lasting
morbidity [38]. Based on study results, 87% of patients who
had recovered from COVID-19 experienced persistence of
at least one symptom (a mean of 60 days after onset of the
rst COVID-19 symptom), particularly dyspnea and fatigue
[39]. However, it should be mentioned that adverse meta-
bolic effects of SARS-CoV-2 infection may be permanent
without specic clinical symptoms.
It is unknown at this time whether diabetes-related
changes that occur in severe COVID-19 will disappear after
the disease has resolved or will be permanent. Patients with
severe COVID-19 could develop high degrees of insulin
resistance that may be asymptomatic. This is consistent with
the SARS virus ndings, causing insulin resistance in 50%
of the patients throughout infection [18]. Follow-up of the
recovered patients with underlying metabolic disorders such
as diabetes would be essential to evaluate developing
microvascular and macrovascular complications in all the
vascular beds, particularly the eye, kidney, and heart [40]. It
is unknown how recovery from the infection does or does
not differ from other forms of severe critical illness [30].
However, based on reports, sepsis recovery experiences
may be applicable to patients recovering from severe
COVID-19 [38]. Receipt of these practices has been related
to lower odds of rehospitalization or death [38]. Taken
together, it is noteworthy that post-recovery sequelae of
COVID-19 may appear months and years after recovery,
and it recommends that for many patients surviving
COVID-19, health care should not end when they are
discharged.
The key role of ACE2 in the interaction between
COVID-19 and diabetes
The interaction between SARS-CoV-2 and angiotensin-
converting enzyme 2 (ACE2) is an essential factor con-
tributing to the new virus to develop different clinical and
metabolic features. ACE2 is expressed in critical metabolic
tissues, including pancreatic beta-cells, adipose tissue, liver,
the small intestine, and the kidneys. It is rational that the
new virus causes pleiotropic alterations in glucose meta-
bolism, which would result in the complication of the
pathophysiology of pre-existing diabetes or the creation of
new mechanisms of disease [11].
ACE2 is the cellular receptor of SARS-CoV-2 and
acts as the door for SARS-CoV-2 to enter host cells.
The cellular serine protease TMPRSS2 is also needed for
ACE2-mediated coronavirus entry into the cells [41]. The
expression of ACE2 in different tissues is proportional to
various symptoms of COVID-19, such as respiratory
symptoms, which are predominant, acute cardiac and kid-
ney injuries, gastrointestinal and liver function abnormal-
ities, and beta-cell damage [4,42]. Understanding the
mechanisms of interaction between the renin-angiotensin
system (RAS) and SARS-CoV-2 can provide adequate
therapy for patients at an increased risk of COVID-19.
ACE2 is a RAS member, which cleaves a single amino acid
residue at the carboxyl terminus of Ang I (Ang 110) and
Ang II (Ang 18) to form Ang 19 and Ang 17, respec-
tively. Vasoconstrictive and inammatory effects of Ang II
are counterbalanced with Ang 17, which has vasodilatory
and anti-inammatory effects [43,44]. The endocytosis of
SARS-CoV-2 results in a reduction in ACE2 activity and a
shift from Ang 17 to Ang II. This alteration may lead to
lung injury, as found in the SARS-CoV infection [45].
Therefore, it should be noted that ACE2 plays different
roles related to COVID-19, so that it is the binding site for
SARS-CoV-2, and on the other hand, its reduced expression
by the virus may involve severe lung injury [44].
Moreover, a higher prevalence of hypokalemia due to
renal potassium wasting is a clinical feature in patients with
COVID-19, which can be explained by the downregulation
of ACE2. This downregulation leads to decreased degra-
dation of Ang II and, consequently, elevated aldosterone
secretion [46]. Also, a decrease in angiotensin 17 and an
Endocrine
increase in angiotensin II concentrations could lead to
altered glucose metabolism in the viruss target cells.
According to a large phenome-wide Mendelian rando-
mization study, T2D was causally associated with increased
expression of ACE2 in the lung. Accordingly, elevated
ACE2 expression due to diabetes and related traits may
inuence COVID-19 risk and exacerbate its complications
[47]. A recent studysndings proposed that patients at old
age, males, and those who have diseases associated with
high expression of ACE2, including hypertension and dia-
betes, may have elevated risk for the delayed clearance of
the virus [48]. It seems that the impacts of diabetes on ACE2
expression depend on the stage of the disease. According to
experimental studies, ACE2 expression increases in early
disease and decreases in later disease [4951]. A study on
mice showed that the lungs ACE2/ACE activity ratio sig-
nicantly decreased in late-stage diabetes [52].
It is noteworthy that soluble ACE2 levels have sig-
nicant associations with all metabolic syndrome compo-
nents, including insulin resistance, obesity, hyperlipidemia,
and hypertension. Intriguingly, APN01, as a recombinant
soluble human ACE2, may have a protective role against
acute lung injury and ARDS induced by SARS-CoV-2. This
drug acts as a decoy receptor and prevents the virus from
entering the cells. APN01 may be the most promising drug
among all medications currently being developed [53]. It
was suggested that the synthetic protease inhibitor camostat,
which blocks TMPRSS2, may prevent and reverse hyper-
insulinemia, hyperglycemia, and hyperlipidemia [54].
Moreover, the interaction between metformin and ACE2
can be important in COVID-19 infection. Metformin acts
through AMP-activated protein kinase (AMPK) activation
and can phosphorylate ACE2. This may result in con-
formational and functional alterations in the receptor,
leading to the decreased binding of the receptor-binding
domain of SARS-CoV-2 to ACE2 [55].
High and aberrantly glycated ACE2 in uncontrolled
hyperglycemia may affect the binding of the spike protein
to ACE2, so resulting in a higher propensity to COVID19
and increased disease severity [56]. It should be noted that
acute and chronic hyperglycemia could have different
impacts on ACE2 expression. While acute hyperglycemia
upregulates ACE2 expression facilitating viral cell entry,
chronic hyperglycemia contributes to downregulating
ACE2 expression, predisposing the cells to become vul-
nerable to the virussinammatory effects. Therefore, both
chronic and acute hyperglycemia can have detrimental
effects [4].
SARS-CoV-2 can also exert direct effects on beta-cells
through ACE2, contributing to exacerbated outcomes in
diabetes patients. The viruss entry into these cells may
cause an acute beta-cell dysfunction that could lead to
uncontrolled hyperglycemia [57,58]. ACE2 was found in
both islets and the exocrine glands. Strikingly, according to
the GTEx database (https://gtexportal.org), the messenger
RNA level of ACE2 was found to be higher in the pancreas
compared with the lung [58], showing that the pancreas is
an essential target of SARS-CoV-2. It was reported that
17% of severe COVID-19 patients had increased levels of
amylase and lipase. The pancreasfocal enlargement or
dilatation of the pancreatic duct was also present in 7.5%
of patients with severe COVID-19 [58]. Direct beta-cell
damage due to SARS-CoV-2 infection could theoretically
cause insulin deciency and autoantigen spread, leading to
severe DKA, insulin dependency, and chronic pancreatic
autoimmunity [4].
ACE2 is expressed on the arterial and venous endothe-
lial cells of the adrenal glands [59]. It was also indicated
that ACE2 and TMPRSS2 are colocalized in adrenocortical
cells. The cortisol concentrations were found to be lower in
critically ill patients with COVID-19 than non-COVID-19
critically ill patients [60]. People with adrenal insufciency
may have a higher rate of respiratory infection-related
deaths, possibly due to impaired immune function and thus
need further care during COVID-19 [59]. The adrenal
glands contribute to the physiology of the stress response
of the organism in health and disease that shows their
important roles in the course of severe COVID-19 infec-
tion. During severe COVID-19, the adrenal glands can be
seriously injured, characterized by the perivascular inl-
tration of CD3+and CD8+T-lymphocytes [61]. Further-
more, adrenal gland disorders such as hyperaldosteronism
and adrenal insufciency can result in carbohydrate
metabolism dysregulation that can affect glucose home-
ostasis [62], and it should be given more attention in the
COVID-19 pandemic.
The importance of pleiotropic alterations in glucose
metabolism
Although SARS-CoV-2 infection is not primarily a meta-
bolic disease, it is found that COVID-19 progression could
be dependent on metabolic mechanisms. Critical conditions,
such as COVID-19, signicantly increase energy demands.
SARS-CoV-2 must rewire cellular metabolism to produce
macromolecules required for replication, assembly, and
existence. Viral proteins elevate extracellular acidication,
which is evidence for direct regulation of glucose metabo-
lism by SARS-CoV-2 in addition to its transcriptional
modulation [41]. Adenosine triphosphate (ATP) is gener-
ated through two related metabolic pathways, glycolysis,
and the tricarboxylic acid (TCA) cycle (Krebscycle) [63].
The imbalance of glucose metabolism could be crucially
contributing to respiratory pathogenic virus infection.
Oxygen supplement at the earliest stage of COVID-19
was proposed to correct the unbalanced glucose aerobic
Endocrine
metabolism [54]. It should be noted that metabolic dys-
function may affect therapies against SARS-CoV-2 infec-
tion because some drugs signicantly depend on ATP to
achieve functionality [64]. Moreover, medicines such as
fenobrate or cloperastine (FDA-approved drugs) that tar-
get host metabolic pathways could play a role in minimizing
virus replication [41].
Like cancer cells, the metabolic milieu was rearranged
in virus-infected cells to facilitate virus production and
replication [65]. In general, viral infection in mammalian
cells shifts cellular metabolism from oxidative phos-
phorylation to glycolysis, which leads to a decrease in
ATP production [66]. Under an anaerobic condition in
COVID-19 patients, on the one hand, the pyruvate pro-
duced from glucose during glycolysis was fermented to
lactate, which leads to the generation of limited amounts of
ATP for urgent biological needs. On the other hand, high
replication of SARS-CoV-2 viruses in infected cells con-
sumes many cellular ATP. As this condition persists (ATP
depletion), lactate delivered to the liver failed to be meta-
bolized through gluconeogenesis, causing increased blood
lactate concentrations in COVID-19 patients [54].
Under the aerobic condition, glucose oxidation can also
be performed through the pentose phosphate pathway in
which nicotinamide adenine dinucleotide phosphate
(NADPH) was produced. NADPH is required to maintain
the proper ratio of oxidized glutathione to glutathione
(GSH), which is crucial for the antioxidant defense system,
contributing to tackling invasive pathogens microorganisms
along with the immune system. Under the persistent
hypoxia state, the depletion of GSH from the pentose
phosphate pathways blockage could induce oxidative
damage during SARS-CoV-2 infection [54,67]. Further-
more, the deciency of glucose-6-phosphate dehy-
drogenase, the key enzyme in the pentose phosphate
pathway, could result in cell apoptosis and inammatory
cytokine release noted in COVID-19 infection [68,69].
Cellular glucose metabolism is hijacked in cells infected
with the SARS-CoV-2 virus [70]. Pro-inammatory cyto-
kines activate immune cells such as macrophages and
dendritic cells to change their metabolism to produce vast
amounts of cytokines. In this metabolic reprogramming,
these cellsability to generate ATP switches from engaging
mitochondrial oxidative phosphorylation to cytosolic
glycolysis. Glycolysis allows pro-inammatory activated
immune cells to generate increased amounts of pro-
inammatory cytokines, including interleukins (IL-1β, IL-
2, IL-6, IL-8), interferons (IFN1αand IFN-1β), and TNF-α
as well as chemokines. These agents can induce excessive
amounts of reactive oxygen and reactive nitrogen species
resulting in extensive nitro-oxidative stress, which involves
respiratory pathophysiology [70,71]. Targeting cytokines
and effectively suppressing the cytokine storm during
SARS-CoV-2 infection could help prevent the deterioration
of COVID-19 patients and reduce mortality [9,72]. How-
ever, it should be noted that therapeutic strategies for tar-
geting the overactive cytokine response must be balanced
with maintaining an adequate inammatory response to
clear the virus [8].
Melatonin forces activated immune cells to abandon
glycolysis in favor of mitochondrial oxidative phosphor-
ylation [70]. Furthermore, this hormone induces Bmal1, a
circadian gene, which inhibits pyruvate dehydrogenase
kinase (PDK) and leads to the disinhibition of the pyruvate
dehydrogenase complex (PDC). PDC catalyzes the mito-
chondrial conversion of pyruvate to acetyl-coenzyme A,
thereby elevating the TCA cycle, oxidative phosphorylation,
and ultimately an increase in ATP production [73]. Intri-
guingly, to counteract quarantine-related sleep disorders, it
was recommended to use food containing or promoting
serotonin and melatonin synthesis at dinner such as leaves,
roots, fruits, and seeds such as almonds, cherries, bananas,
and oats along with increased physical activity [74].
PDC links glycolysis to TCA and fatty acid synthesis
and can be a crucial modulator of energy and metabolic
homeostasis. Ang II downregulates the activity of PDC
through its phosphorylation and acetylation. The reduced
PDC can decrease the rate of glycolysis, leading to an
uncoupling between glycolysis and TCA, which would
induce intracellular acidosis and energy depletion [66].
Insulin resistance is related to elevated skeletal muscle PDK
contributing to the phosphorylation and inactivation of
PDC. It is noteworthy that dichloroacetate, as a PDK4
inhibitor, could restore PDC activity and ATP level,
improve metabolism disorders, suppress cytokine storm and
viral replication [75].
The key rate-limiting enzymes in glycolysis, including
pyruvate kinase isozyme and hexokinase 2, were upregu-
lated in SARS-CoV-2 infection [41]. The conversion of
phosphoenolpyruvate to pyruvate is a crucial step in gly-
colysis, which is catalyzed by PK. PKM1 and PKM2 are the
two major isoenzymes of PK. PKM1 is the more enzyma-
tically active form and contributes to the entry of pyruvate
into TCA. PKM2 exhibits lower enzymatic activity than
PKM1 and involves elevated glycolysis, leading to the
cytosolic accumulation of lactate and other metabolic inter-
mediates [63]. Cytosolic lactate levels and lactate to pyr-
uvate ratio were increased in COVID-19 patients requiring
ICU admission compared with healthy controls, showing
that the increase in lactate can be due to a fundamental
metabolic shift rather than merely an enhancement in overall
metabolism [63]. It was also reported that lactate dehy-
drogenase levels were moderately higher in the deceased
than in survival elderly patients with COVID-19 [76].
Inactivated immune cells, decreased TCA could lead
to the accumulation of several metabolites, including
Endocrine
succinate, which is a crucial TCA cycle intermediate. As an
endogenous danger signal, Succinate contributes to stabi-
lizing hypoxia-inducible factor-1a, which increases the
expression of IL-1ß during inammation [76]. It was indi-
cated that the dysregulated TCA and the upregulated gly-
colysis were mediated by the inammatory transcription
factors NF-кB and RELA [41]. In a multi-center study on
elderly patients with COVID-19, immune-related metabolic
index (an index based on the ux of corresponding meta-
bolic reactions) was found as a potential risk factor (odds
ratio: 6.42 [95% CI 2.6615.48], p< 0.001) [76].
After entering the human cells, SARS-CoV-2 increases
the hexosamine biosynthetic pathway (HBP) to secure
excessive glucose consumption and substrates for rapid
replication. This enhanced HBP would result in an
excess of the enzyme O-GlcNAc (N- acetylglucosamine)
transferase (OGT), which would, in turn, trigger massive
amounts of interferon regulatory factor-5 (IRF5). IRF5
plays a role as a mediator in the induction of pro-
inammatory cytokines such as IL-6, IL-12, IL-23, and
TNF-α, is contributed to the recruitment of inammatory
genes with NF-кB and in determining the inammatory
macrophage phenotype. IRF5 and OGT contribute to
worsening the IкBkinase-NF-кBpro-inammatory path-
way resulting in a tremendous inammatory cytokine gene
overexpression prole and deleterious endoplasmic reti-
culum stress that ultimately lead to hyperinammation, a
cytokine storm, and multiorgan failure (Fig. 1)[65]. Fur-
thermore, SARS-CoV-2, through this mechanism, may
increase vascular complications in patients with diabetes,
and it may also explain the sudden cardiac deaths in
COVID-19 [77].
Glucose
Glucose-6p
Fructose-6p
NADPH
GSH
Antioxidant defense
system
Hexosamine biosynthetic
pathway
IRF5
Proinflammatory
pathways
HK2
Pentose phosphate
pathway
Pyruvate
PKM2
LDH
Lactate
PEP
PDC PDK
Pyruvate
Acetyl-CoA
TCA
cycle
Fig. 1 SARS-CoV-2-induced
alterations in glucose
metabolism. In mammalian
cells, SARS-CoV-2 shifts
cellular metabolism from
oxidative phosphorylation to
glycolysis, which results in a
decrease in ATP generation. The
key rate-limiting enzymes in
glycolysis, including PKM2 and
HK2, were upregulated in
COVID-19. The decreased PDC
can lead to an uncoupling
between glycolysis and TCA,
which would induce intracellular
acidosis and energy depletion.
Under an anaerobic condition in
COVID-19, the depletion of
GSH from the pentose
phosphate pathways blockage
could cause oxidative damage
by SARS-CoV-2. The new virus
elevates the hexosamine
biosynthetic pathway, which
would result in massive amounts
of IRF5. IRF5 involves
worsening pro-inammatory
pathways that ultimately lead to
hyper inammation, a cytokine
storm, and multiorgan failure.
GSH, glutathione; HK2,
hexokinase 2; IRF5, interferon
regulatory factor-5; LDH, lactate
dehydrogenase; NADPH,
nicotinamide adenine
dinucleotide phosphate; PDC,
pyruvate dehydrogenase
complex; PEP,
phosphoenolpyruvate; PDK,
pyruvate dehydrogenase kinase;
PKM2, pyruvate kinase isozyme
M2; TCA, tricarboxylic acid
Endocrine
Conclusion
Since the pathophysiology of COVID-19 is still mostly
unknown, identifying the metabolic mechanisms contribut-
ing to its progression is essential to provide specic ways to
prevent and improve this dangerous viruss detrimental
effects. It is also necessary to understand the long-term
consequences that will affect the health of patients with
diabetes who survived COVID-19. SARS-CoV-2-induced
metabolic alterations in immune cells can result in hyper
inammation and a cytokine storm. These alterations could
prove useful therapeutic targets for combating COVID-19.
The ndings show that the new virus may induce new-onset
diabetes with uncertain metabolic and clinical features,
supporting a potential role of COVID-19 in the develop-
ment of diabetes.
Compliance with ethical standards
Conict of interest The authors declare no competing interests.
Publishers note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional afliations.
References
1. C. Covid, C. COVID, C. COVID, N. Chow, K. Fleming-Dutra, R.
Gierke et al. Preliminary estimates of the prevalence of selected
underlying health conditions among patients with coronavirus
disease 2019United States, February 12March 28, 2020.
Morbidity Mortal. Wkly. Rep. 69(13), 382 (2020)
2. E. Maddaloni, L. DOnofrio, F. Alessandri, C. Mignogna, G. Leto,
G. Pascarella et al. Cardiometabolic multimorbidity is associated
with a worse Covid-19 prognosis than individual cardiometabolic
risk factors: a multicentre retrospective study (CoViDiab II).
Cardiovasc. Diabetol. 19(1), 111 (2020)
3. B. Li, J. Yang, F. Zhao, L. Zhi, X. Wang, L. Liu et al. Prevalence
and impact of cardiovascular metabolic diseases on COVID-19 in
China. Clin. Res. Cardiol. 109(5), 531538 (2020)
4. S.R. Bornstein, F. Rubino, K. Khunti, G. Mingrone, D. Hopkins,
A.L. Birkenfeld et al. Practical recommendations for the man-
agement of diabetes in patients with COVID-19. Lancet Diabetes
Endocrinol. 8, 546550 (2020)
5. S. Mirmohammadi, A. Kianmehr, M. Are, A. Mahrooz, Bio-
chemical parameters and pathogenesis of SARS-CoV-2 infection
in vital organs: COVID-19 outbreak in Iran. N. Microbes N.
Infect. 38, 100792 (2020)
6. L. Shang, M. Shao, Q. Guo, J. Shi, Y. Zhao, J. Xiaokereti et al.
Diabetes mellitus is associated with severe infection and mortality
in patients with COVID-19: a systematic review and meta-
analysis. Arch. Med. Res. 51, 700709 (2020)
7. A. De Lorenzo, S. Escobar, E. Tibiriçá, Systemic endothelial
dysfunction: a common pathway for COVID-19, cardiovascular
and metabolic diseases. Nutr., Metab. Cardiovasc. Dis. 30(8),
14011402 (2020)
8. R.J. Jose, A. Manuel, COVID-19 cytokine storm: the interplay
between inammation and coagulation. Lancet Respir. Med. 8(6),
e46e47 (2020)
9. D. Ragab, H. Salah Eldin, M. Taeimah, R. Khattab, R. Salem, The
COVID-19 cytokine storm; what we know so far. Front. Immunol.
11, 1446 (2020)
10. A. Ceriello, V. De Nigris, F. Prattichizzo, Why is hyperglycaemia
worsening COVID19 and its prognosis? Diabetes, Obes. Metab.
22(10), 19511952 (2020)
11. S. Cuschieri, S. Grech, COVID-19 and diabetes: the why, the
what and the how. J. Diabetes Complicat. 34, 107637 (2020)
12. L. Zhu, Z.-G. She, X. Cheng, J.-J. Qin, X.-J. Zhang, J. Cai et al.
Association of blood glucose control and outcomes in patients
with COVID-19 and pre-existing type 2 diabetes. Cell Metab. 31,
10681077 (2020)
13. A. Wang, W. Zhao, Z. Xu, J. Gu, Timely blood glucose man-
agement for the outbreak of 2019 novel coronavirus disease
(COVID-19) is urgently needed. Diabetes Res. Clin. Pract. 162,
108118 (2020)
14. England N., Improvement N. After-care needs of inpatients
recovering from COVID-19. 2020;001559
15. J. Li, X. Wang, J. Chen, X. Zuo, H. Zhang, A. Deng, COVID19
infection may cause ketosis and ketoacidosis. Diabetes, Obes.
Metab. 22, 19351941 (2020)
16. C. Kamrath, K. Mönkemöller, T. Biester, T.R. Rohrer, K.
Warncke, J. Hammersen et al. Ketoacidosis in children and ado-
lescents with newly diagnosed type 1 diabetes during the COVID-
19 pandemic in Germany. JAMA 324, 801804 (2020)
17. E. Maddaloni, R. Buzzetti, Covid-19 and diabetes mellitus:
unveiling the interaction of two pandemics. Diabetes/Metab. Res.
Rev 36, e33213321 (2020)
18. L. Marchand, M. Pecquet, C. Luyton, Type 1 diabetes onset trig-
gered by COVID-19. Acta Diabetologica 57,12651266 (2020)
19. S. Smith, A. Boppana, J. A. Traupman, E. Unson, D. A. Maddock,
K. Y. Chao, et al. Impaired glucose metabolism in patients with
diabetes, prediabetes and obesity is associated with severe Covid-
19. J. Med. Virol. 93, 409415 (2021)
20. A. Hussain, B. Bhowmik, N.C. do Vale Moreira, COVID-19 and
diabetes: Knowledge in progress. Diabetes Res. Clin. Pract. 162,
108142 (2020)
21. A. Yehya, S. Carbone, Managing type 2 diabetes mellitus during
COVID-19 pandemic: the bittersweet. Diabetes/Metab Res Rev
37, e3360 (2021)
22. S. Knapp, Diabetes and infection: Is there a link?A mini-
review. Gerontology 59(2), 99104 (2013)
23. G. Daryabor, M.R. Atashzar, D. Kabelitz, S. Meri, K. Kalantar,
The effects of type 2 diabetes mellitus on organ metabolism and
the immune system. Front. Immunol. 11, 1582 (2020)
24. M.I. Maiorino, G. Bellastella, D. Giugliano, K. Esposito, Cooling
down inammation in type 2 diabetes: how strong is the evidence
for cardiometabolic benet? Endocrine 55(2), 360365 (2017)
25. A. Berbudi, N. Rahmadika, A. Cahyadi, R. Ruslami, Type 2
Diabetes and its Impact on the Immune System. Curr. diabetes
Rev. 16(5), 442 (2020)
26. M. Delamaire, D. Maugendre, M. Moreno, M.C. Le Goff, H.
Allannic, B. Genetet, Impaired leucocyte functions in diabetic
patients. Diabet. Med. 14(1), 2934 (1997)
27. S.N. Zykova, T.G. Jenssen, M. Berdal, R. Olsen, R. Myklebust, R.
Seljelid, Altered cytokine and nitric oxide secretion in vitro by
macrophages from diabetic type II-like db/db mice. Diabetes 49
(9), 14511458 (2000)
28. G. Patti, I. Cavallari, F. Andreotti, P. Calabro, P. Cirillo, G. Denas
et al. Prevention of atherothrombotic events in patients with dia-
betes mellitus: from antithrombotic therapies to new-generation
glucose-lowering drugs. Nat. Rev. Cardiol. 16(2), 113130 (2019)
29. W. Guo, M. Li, Y. Dong, H. Zhou, Z. Zhang, C. Tian, et al.
Diabetes is a risk factor for the progression and prognosis of
COVID-19. Diabetes/Metab. Res. Rev 36, e3319 (2020)
Endocrine
30. L.D.J. Bos, D. Brodie, C.S. Calfee, Severe COVID-19 infections
knowledge gained and remaining questions. JAMA Intern.
Med. 181,911 (2021)
31. H.R. Murphy, Managing diabetes in pregnancy before, during, and
after COVID-19. Diabetes Technol. Ther. 22(6), 454461 (2020)
32. T.E. van Gemert, R.G. Moses, A.V. Pape, G.J. Morris, Gestational
diabetes mellitus testing in the COVID19 pandemic: the pro-
blems with simplifying the diagnostic process. Aust. N.Z. J.
Obstet. Gynaecol. 60(5), 671674 (2020)
33. D. Di Mascio, A. Khalil, G. Saccone, G. Rizzo, D. Buca, M.
Liberati et al. Outcome of coronavirus spectrum infections
(SARS, MERS, COVID-19) during pregnancy: a systematic
review and meta-analysis. Am. J. Obstet. Gynecol. 2(2), 100107
(2020)
34. S. Smati, P. M. Moreau, A. Bourdiol, S. Ploteau, S. Hadjadj, B.
Cariou, Euglycaemic ketoacidosis during gestational diabetes with
concomitant COVID-19 infection. Diabetes Metab. 47, 101181
(2021)
35. A.K. Singh, R. Singh, Hyperglycemia without diabetes and new-
onset diabetes are both associated with poorer outcomes in
COVID-19. Diabetes Res. Clin. Pract. 167, 108382 (2020)
36. J. Zhou, J. Tan, Diabetes patients with COVID-19 need better
blood glucose management in Wuhan, China. Metabolism 107,
154216 (2020)
37. Q. Wu, L. Zhou, X. Sun, Z. Yan, C. Hu, J. Wu et al. Altered lipid
metabolism in recovered SARS patients twelve years after infec-
tion. Sci. Rep. 7(1), 112 (2017)
38. H.C. Prescott, T.D. Girard, Recovery From Severe COVID-19:
leveraging the Lessons of Survival From Sepsis. JAMA 324(8),
739740 (2020)
39. A. Carfì, R. Bernabei, F. Landi, Persistent symptoms in patients
after acute COVID-19. JAMA 324, 603605 (2020)
40. R. Dalan, B. O. Boehm, The implications of COVID-19 infection
on the endothelium: a metabolic vascular perspective. Diabetes/
Metab. Res. Rev 37, e3402 (2021)
41. Ehrlich A., Uhl S., Ioannidis K., Hofree M., tenOever B. R.,
Nahmias Y. The SARS-CoV-2 transcriptional metabolic signature
in lung epithelium. Available at SSRN 3650499
42. M. Gheblawi, K. Wang, A. Viveiros, Q. Nguyen, J.-C. Zhong, A.
J. Turner et al. Angiotensin-converting enzyme 2: SARS-CoV-2
receptor and regulator of the renin-angiotensin system: celebrating
the 20th anniversary of the discovery of ACE2. Circ. Res. 126
(10), 14561474 (2020)
43. C.M. Ferrario, A.J. Trask, J.A. Jessup, Advances in biochemical
and functional roles of angiotensin-converting enzyme 2 and
angiotensin-(17) in regulation of cardiovascular function. Am. J.
Physiol.Heart Circulatory Physiol. 289(6), H2281H2290 (2005)
44. M. AlGhatrif, O. Cingolani, E.G. Lakatta, The dilemma of cor-
onavirus disease 2019, aging, and cardiovascular disease: insights
from cardiovascular aging science. JAMA Cardiol. 5, 747748
(2020)
45. K. Kuba, Y. Imai, S. Rao, H. Gao, F. Guo, B. Guan et al. A crucial
role of angiotensin converting enzyme 2 (ACE2) in SARS
coronavirusinduced lung injury. Nat. Med. 11(8), 875879 (2005)
46. Li X., Hu C., Su F., Dai J. Hypokalemia and clinical implications
in patients with coronavirus disease 2019 (COVID-19). MedRxiv.
2020
47. S. Rao, A. Lau, H.-C. So, Exploring diseases/traits and blood
proteins causally related to expression of ACE2, the putative
receptor of SARS-CoV-2: A Mendelian Randomization analysis
highlights tentative relevance of diabetes-related traits. Diabetes
Care. 43, 14161426 (2020)
48. Chen X., Hu W., Ling J., Mo P., Zhang Y., Jiang Q., et al.
Hypertension and diabetes delay the viral clearance in COVID-19
patients. medRxiv. 2020
49. S.M. Bindom, E. Lazartigues, The sweeter side of ACE2: phy-
siological evidence for a role in diabetes. Mol. Cell. Endocrinol.
302(2), 193202 (2009)
50. M. Ye, J. Wysocki, P. Naaz, M.R. Salabat, M.S. LaPointe, D.
Batlle, A.C.E. Increased, 2 and decreased ACE protein in renal
tubules from diabetic mice: a renoprotective combination?
Hypertension 43(5), 11201125 (2004)
51. J. Wysocki, M. Ye, M.J. Soler, S.B. Gurley, H.D. Xiao, K.E.
Bernstein et al. ACE and ACE2 activity in diabetic mice. Diabetes
55(7), 21322139 (2006)
52. H. Roca-Ho, M. Riera, V. Palau, J. Pascual, M.J. Soler, Char-
acterization of ACE and ACE2 expression within different organs
of the NOD mouse. Int. J. Mol. Sci. 18(3), 563 (2017)
53. A.M. Angelidi, M.J. Belanger, C.S. Mantzoros, Commentary:
COVID-19 and diabetes mellitus: what we know, how our patients
should be treated now, and what should happen next. Metabolism
107, 154245 (2020)
54. Z. Li, G. Liu, L. Wang, Y. Liang, Q. Zhou, F. Wu et al. From the
insight of glucose metabolism disorder: Oxygen therapy and
blood glucose monitoring are crucial for quarantined COVID-19
patients. Ecotoxicol. Environ. Saf. 197, 110614 (2020)
55. S. Sharma, A. Ray, B. Sadasivam, Metformin in COVID-19: a
possible role beyond diabetes. Diabetes Res. Clin. Pract. 164,
108183 (2020)
56. A. Brufsky, Hyperglycemia, hydroxychloroquine, and the
COVID19 pandemic. J. Med. Virol. 92(7), 770775 (2020)
57. G. Lisco, A. De Tullio, V.A. Giagulli, E. Guastamacchia, G. De
Pergola, V. Triggiani, Hypothesized mechanisms explaining poor
prognosis in type 2 diabetes patients with COVID-19: a review.
Endocrine 70, 441453 (2020)
58. F. Liu, X. Long, B. Zhang, W. Zhang, X. Chen, Z. Zhang, ACE2
expression in pancreas may cause pancreatic damage after SARS-
CoV-2 infection. Clin. Gastroenterol. Hepatol. 18(9), 21282130
(2020)
59. R. Pal, COVID-19, hypothalamo-pituitary-adrenal axis and clin-
ical implications. Endocrine 68(2), 251252 (2020)
60. Y. Mao, B. Xu, W. Guan, D. Xu, F. Li, R. Ren et al. The adrenal
cortex, an underestimated site of SARS-CoV-2 infection. Front.
Endocrinol. 11, 593179 (2020)
61. V.A. Zinserling, N.Y. Semenova, A.G. Markov, O.V. Rybal-
chenko, J. Wang, R.N. Rodionov et al. Inammatory cell inl-
tration of adrenals in COVID-19. Horm. Metab. Res. 52(09),
639641 (2020)
62. K.J. Ioakim, G.I. Sydney, S.A. Paschou, Glucose metabolism
disorders in patients with adrenal gland disorders: pathophysiol-
ogy and management. Hormones 19(2), 135143 (2020)
63. O.J. McElvaney, N. McEvoy, O.F. McElvaney, T.P. Carroll, M.P.
Murphy, D.M. Dunlea et al. Characterization of the inammatory
response to severe COVID-19 illness. Am. J. Respir. Crit. Care
Med. 202, 812821 (2020)
64. M. Migaud, S. Gandotra, H.S. Chand, M.N. Gillespie, V.J.
Thannickal, R.J. Langley, Metabolomics to predict antiviral drug
efcacy in COVID-19. Am. J. Respiratory Cell Mol. Biol. 63(3),
396398 (2020)
65. H.A. Laviada-Molina, I. Leal-Berumen, E. Rodriguez-Ayala, R.A.
Bastarrachea, Working hypothesis for glucose metabolism and
SARS-CoV-2 replication: interplay between the hexosamine
pathway and interferon RF5 triggering hyperinammation. Role
of BCG vaccine? Front. Endocrinol. 11, 514 (2020)
66. J. Mori, G. Y. Oudit, G. D. Lopaschuk, SARS-CoV-2 perturbs the
Renin-Angiotensin System and energy metabolism. Am. J. Phy-
siol.-Endocrinol. Metab. 319, E43E47 (2020)
67. E. Eshak, A. Arafa, Thiamine deciency and cardiovascular
disorders. Nutr., Metab. Cardiovasc Dis. 28(10), 965972
(2018)
Endocrine
68. M. Çiftçi, İ. Özmen, M.E. Büyükokuroğlu, S. Pençe, Küfrevioğlu
Öİ. Effects of metamizol and magnesium sulfate on enzyme
activity of glucose 6-phosphate dehydrogenase from human ery-
throcyte in vitro and rat erythrocyte in vivo. Clin. Biochem. 34(4),
297302 (2001)
69. S.F. Nabavi, S. Habtemariam, A. Sureda, M. Banach, I. Berindan-
Neagoe, C.A. Cismaru et al. Glucose-6-phosphate dehydrogenase
deciency and SARS-CoV-2 mortality: Is there a link and what
should we do? Clin. Biochem. 86,3133 (2020)
70. R.J. Reiter, R. Sharma, Q. Ma, C. Liu, W. Manucha, P. Abreu-
Gonzalez et al. Plasticity of glucose metabolism in activated
immune cells: advantages for melatonin inhibition of COVID-19
disease. Melatonin Res. 3(3), 362379 (2020)
71. M.A. El-Missiry, Z.M. El-Missiry, Melatonin is a potential adju-
vant to improve clinical outcomes in individuals with obesity and
diabetes with coexistence of Covid-19. Eur. J. Pharmacol. 882,
173329 (2020)
72. Q. Ye, B. Wang, J. Mao, Cytokine storm in COVID-19 and
treatment. J. Infect. 80(6), 607613 (2020)
73. G. Anderson, R.J. Reiter, Melatonin: roles in inuenza, Covid19,
and other viral infections. Rev. Med. Virol. 30(3), e2109 (2020)
74. L. Barrea, G. Pugliese, L. Framondi, R. Di Matteo, D. Laudisio, S.
Savastano, et al. Does Sars-Cov-2 threaten our dreams? Effect of
quarantine on sleep quality and body mass index. J Transl Med.
18, 318 (2020)
75. L. Luzi, M.G. Radaelli, Inuenza and obesity: its odd relationship
and the lessons for COVID-19 pandemic. Acta Diabetol. 57,
759764 (2020)
76. G. Tannahill, A. Curtis, J. Adamik, E. Palsson-McDermott, A.
McGettrick, G. Goel et al. Succinate is an inammatory signal that
induces IL-1βthrough HIF-1α.Nature496(7444), 238242 (2013)
77. E. Akkus, M. Sahin, Related molecular mechanisms of COVID-
19, hypertension, and diabetes. Am. J. Physiol.-Endocrinol.
Metab. 318(6), E881E881 (2020)
Endocrine
... Відомо, що пацієнти з ЦД 2 типу не лише схильні до інфекцій, спричинених бактеріями, грибами та вірусами, а і мають тяжчий перебіг захворювання порівняно з пацієнтами без діабету. Триває вивчення механізмів, які спричиняють зростання смертності та ризику ускладнень при захворюваннях легень у пацієнтів із ЦД 2 ти пу [12,15,18,20,26,27]. Установлено, що хронічний запальний процес та порушення метаболізму глюкози при ЦД 2 типу змінюють регуляцію імунної системи (порушення функції Т-клітин, макрофагів і хемотаксис нейтрофілів, що призводить до інфікування) [12,14,20]. ...
... Триває вивчення механізмів, які спричиняють зростання смертності та ризику ускладнень при захворюваннях легень у пацієнтів із ЦД 2 ти пу [12,15,18,20,26,27]. Установлено, що хронічний запальний процес та порушення метаболізму глюкози при ЦД 2 типу змінюють регуляцію імунної системи (порушення функції Т-клітин, макрофагів і хемотаксис нейтрофілів, що призводить до інфікування) [12,14,20]. Резистентність до інсуліну та зростання рівня цитокінів спричиняють запальну реакцію і зростання окисного стресу в легенях. ...
... Неферментативне глікозилювання робить колаген менш сприйнятливим до протеолізу, що призводить до його накопичення в сполучній тканині легень і спричиняє рестриктивне захворювання. Гіперглікемія також знижує мукоциліарний кліренс, що може призвести до посилення легеневої інфекції [15,16,20]. На відміну від морфофункціональних порушень енергетичні розлади в цих пацієнтів значно менше досліджені [9]. ...
Article
Full-text available
Objective — to characterize ultrastructural changes in lung and liver cells associated with disturbances in energy metabolism during the comorbid course of inflammation in the lungs and type 2 diabetes mellitus (T2DM). Materials and methods. The study was conducted on adult Wistar rats, divided into four groups: 1) control; 2) rats with simulated inflammation by lipopolysaccharide (LPS) administration (0.5 mg/kg intraperitoneally); 3) rats with insulin resistance induced by a high-fat diet (58 % of total calorie intake from lipids) and LPS injection on the 24th day of the experiment; 4) rats with simulated T2DM by a high-fat diet, streptozotocin injection (25 mg/kg body weight) on the 14th day, and LPS injection on the 24th day of the experiment. The ultrastructure of lung and liver cells was evaluated by electron microscopy in tissue samples obtained from anesthetized animals three days after LPS administration. Results and discussion. In the simulation of inflammation induced by LPS-mediated cytokine mechanisms, inflam­matory symptoms were observed in the lungs, with alveolar type II cells showing mitochondrial dysfunc­tion, a large number of free and attached ribosomes, indicating disruptions in energy metabolism, and the development of compensatory-adaptive reactions (intensification of protein synthesis, reparative processes in mitochondria and their interaction with the endoplasmic reticulum). In the comorbid course of LPS-in­duced inflammation in the lungs and T2DM, enhanced inflammatory manifestations in the lungs were registered against the background of lipid infiltration, increased aero-hydrate barrier hydration, endothelial dysfunction and signs of mitochondrial dysfunction in alveolar type II cells. Compensatory protein synthesis increased, indicating the intensification of alteration processes, and worsening of energy and lipid metabolism. In the simulation of LPS-induced inflammation in the lungs with insulin resistance, pathological manifestations were less pronounced, and mitochondrial dysfunction was not observed, indicating the maintenance of energy metabolism under these conditions, with insulin resistance playing a partial role in the unfavourable course of comorbid pathology. In the liver, during the comorbid course of LPS-induced inflammation in the lungs and T2DM, increased steatosis, endothelial dysfunction, signs of mitochondrial dysfunction in hepatocytes, pronounced compensatory protein synthesis in bound ribosomes, and the formation of endoplasmic reticulum-associated membranes with mitochondria were detected. Conclusions. During the comorbid course of LPS-induced lung inflammation and T2DM, disruptions in energy metabolism intensify, mediated by respiratory and mitochondrial dysfunction, and compensatory protein synthesis and interaction of mitochondria with the endoplasmic reticulum are enhanced. The results suggest that the pathogenesis of comorbid inflammation in the lungs and T2DM or insulin resistance occurs through possible compensation of carbohydrate and energy metabolism disorders at the expense of worsening lipid metabolism and progression of obesity.
... In T2DM, elevated IL-17 levels are linked to adipose tissue inflammation, regulating proinflammatory cytokines, and contributing to insulin resistance [86]. The cytokine storm induced by SARS-CoV-2 in diabetic patients exacerbates the systemic immune imbalance, potentially worsening their clinical condition [87,88]. ...
Article
Full-text available
Post-COVID syndrome, also known as long COVID, includes a range of symptoms that persist for months or even years after initial infection such as fatigue, shortness of breath, joint pain, chest pain, muscle aches, and heart palpitations, among others. In addition, long COVID is related with new-onset diseases such as diabetes mellitus. The association between SARS-CoV-2 infections and the development of diabetes mellitus is complex and not fully understood. Therefore, the objective of this article was to summarize the state of the art in possible mechanisms involved in the development of diabetes mellitus in the post-COVID-19 era, particularly the impact of SARS-CoV-2 variants on molecular mimicry, the role of viral m6A RNA methylation, and the potential associations between these factors. A better understanding of the combinatorial effects of these mechanisms is paramount for both clinicians and researchers alike because it could help tailor more effective treatment strategies, enhance patient care, and guide future research efforts.
... A more profound knowledge of the pathophysiology of COVID-19, with the identification of metabolic mechanisms, is essential to provide specific ways to prevent and improve the consequences of SARS-CoV-2 infection. 46,47 A significant increase in glycemic values was observed in patients with previously good glycemic control and COVID-19 infection, needing treatment with insulin therapy in higher doses. This suggests the possibility of pancreatic invasion by SARS-CoV-2. ...
Article
Full-text available
Purpose Patients with diabetes mellitus (DM) are more susceptible to viral and bacterial infections, facing a more severe prognosis and higher mortality rates. The study’s main aim was to evaluate the survival and mortality rates of patients with type 2 diabetes (T2DM) and SARS-CoV-2 virus infection alongside the main factors influencing the prognosis. Patients and Methods The present study included 186 patients with T2DM and SARS-CoV-2 virus infection admitted to the COVID-19 Department of the “Pius Brînzeu” Emergency Clinical County University Hospital between November 2020 and March 2021. Patients had investigations performed upon arrival in the emergency room and during hospitalization. We analyzed the risk of negative prognosis based on clinical data (oxygen saturation (SatO2), respiratory rate (RR), lung damage), glycemic control (HbA1c, glycemia at hospital admission), and the duration of T2DM. Results The mortality rate in the studied group was 36.6%. All deceased patients had previously been diagnosed with hypertension; 95.58% had a body mass index (BMI) greater than 25 kg/m², and 79.41% presented with cardiovascular disease (CVD). Compared to those who recovered, statistically significant differences were observed in BMI, glycemic levels at admission, glycosylated hemoglobin levels (HbA1c), SatO2, RR, and lung damage. Valid statistically significant predictors for death in T2DM patients with COVID-19 were hyperglycemia at admission > 198mg/dl, HbA1c> 8.6%, and SatO2≤ 87%. Conclusion SatO2, glycemia at hospital admission, and HbA1c had the highest sensitivity and specificity to predict the prognosis of T2DM patients with SARS-CoV-2 infection. Glycemic control is essential in the prognosis of patients with DM and COVID-19 infection. The prognosis was worse if other comorbidities were associated, especially hypertension and CVD.
... These conditions are characterized by metabolic dysregulation and chronic inflammation, which can exacerbate the severity of SARS-CoV-2 infection [38,109]. In addition to increasing the prevalence of fractures [110], diabetes may also affect the efficacy of therapies against SARS-CoV-2 infection [111] and cause increased inflammation and a protracted course of infection and recovery. For SARS-CoV-2 infection survivors, a systematic review found a 64% greater risk of new onset diabetes when comparing patients with SARS-CoV-2 infection to patients without it [112]. ...
Article
Full-text available
Purpose of Review SARS-CoV-2 infection, the culprit of the COVID-19 pandemic, has been associated with significant long-term effects on various organ systems, including bone health. This review explores the current understanding of the impacts of SARS-CoV-2 infection on bone health and its potential long-term consequences. Recent Findings As part of the post-acute sequelae of SARS-CoV-2 infection, bone health changes are affected by COVID-19 both directly and indirectly, with multiple potential mechanisms and risk factors involved. In vitro and preclinical studies suggest that SARS-CoV-2 may directly infect bone marrow cells, leading to alterations in bone structure and osteoclast numbers. The virus can also trigger a robust inflammatory response, often referred to as a "cytokine storm", which can stimulate osteoclast activity and contribute to bone loss. Clinical evidence suggests that SARS-CoV-2 may lead to hypocalcemia, altered bone turnover markers, and a high prevalence of vertebral fractures. Furthermore, disease severity has been correlated with a decrease in bone mineral density. Indirect effects of SARS-CoV-2 on bone health, mediated through muscle weakness, mechanical unloading, nutritional deficiencies, and corticosteroid use, also contribute to the long-term consequences. The interplay of concurrent conditions such as diabetes, obesity, and kidney dysfunction with SARS-CoV-2 infection further complicates the disease's impact on bone health. Summary SARS-CoV-2 infection directly and indirectly affects bone health, leading to potential long-term consequences. This review article is part of a series of multiple manuscripts designed to determine the utility of using artificial intelligence for writing scientific reviews.
... Early studies from China identified diabetes as a risk factor for severe COVID-19 syndrome, but because of the difficulty clinically defining obesity; the two diagnoses may be combined [6,137]. Diabetes has been shown in multiple studies to increase the severity and mortality associated with SARS-CoV-2 infection [138,139]. The association of obesity and insulin resistance is a well-recognized phenomenon [140]. ...
Article
Full-text available
Purpose of Review SARS-CoV-2 drove the catastrophic global phenomenon of the COVID-19 pandemic resulting in a multitude of systemic health issues, including bone loss. The purpose of this review is to summarize recent findings related to bone loss and potential mechanisms. Recent Findings The early clinical evidence indicates an increase in vertebral fractures, hypocalcemia, vitamin D deficiencies, and a loss in BMD among COVID-19 patients. Additionally, lower BMD is associated with more severe SARS-CoV-2 infection. Preclinical models have shown bone loss and increased osteoclastogenesis. The bone loss associated with SARS-CoV-2 infection could be the result of many factors that directly affect the bone such as higher inflammation, activation of the NLRP3 inflammasome, recruitment of Th17 cells, the hypoxic environment, and changes in RANKL/OPG signaling. Additionally, SARS-CoV-2 infection can exert indirect effects on the skeleton, as mechanical unloading may occur with severe disease (e.g., bed rest) or with BMI loss and muscle wasting that has also been shown to occur with SARS-CoV-2 infection. Muscle wasting can also cause systemic issues that may influence the bone. Medications used to treat SARS-CoV-2 infection also have a negative effect on the bone. Lastly, SARS-CoV-2 infection may also worsen conditions such as diabetes and negatively affect kidney function, all of which could contribute to bone loss and increased fracture risk. Summary SARS-CoV-2 can negatively affect the bone through multiple direct and indirect mechanisms. Future work will be needed to determine what patient populations are at risk of COVID-19-related increases in fracture risk, the mechanisms behind bone loss, and therapeutic options. This review article is part of a series of multiple manuscripts designed to determine the utility of using artificial intelligence for writing scientific reviews.
... The more severe the disease, the greater the increase in antibody levels. Under the mediation of the reaction, the secretion of insulin decreases, which will affect the thyroid function, thereby increasing the content of antibody, aggravating the degree of systemic disease, which is not conducive to the normal life [17,18]. The previous study has shown that the higher level of HbA1c indicates the higher TGAb level in diabetic patients [19]. ...
Article
Full-text available
The association of thyroid hormone antibodies and glycolipid metabolism indicators with Type 2 diabetes mellitus (T2DM) was explored. As the disease worsens, the levels of thyroglobulin antibody (TGAb), thyroid peroxidase antibody (TPOAb), and thyroid-stimulating hormone (TSH) was increased, and the levels of total tri-iodothyronine (TT3) and total thyroxine (TT4) was decreased (P < 0.001). The severe, medium, and light group had higher level of high-density lipoprotein (HDL), lower level of total cholesterol (TC), low-density lipoprotein (LDL), glycosylated hemoglobin (HbA1c), triacylglycerol (TAG), and fasting blood sugar (FBG) than the control group (P < 0.05). The level of HDL was lower in the severe group than the light group and the medium group, but the levels of TC, LDL, HbA1c, TAG, and FBG were increased with the progress of T2DM (P < 0.001). The levels of TGAb, TPOAb, and TSH in patients with T2DM were positively correlated with the levels of TC, LDL, HbA1c, TAG, and FBG (P < 0.05), and were negatively correlated with HDL levels (P < 0.05). The life quality score was lower in the severe group than the light and the medium group (P < 0.001). Among the above indicators, the predictive value of TT3, TT4, and HbA1c in T2DM was better. Clinically, detecting the levels of thyroid hormone antibodies and glycolipid metabolism indicators had a certain predictive value for the severity of T2DM. Main findings: The results of this study found that the thyroid hormone antibody and glycolipid metabolism levels in T2DM patients were abnormal, and had different degrees of impact on the quality of life of patients. Thus, monitoring these indicators had certain predictive value for the severity of the disease, and also had a certain degree of suggestive effect on the evaluation of diabetic vascular complications. Clinically, attention should be paid to the screening of thyroid disease in diabetic patients, and the assessment and prognosis of thyroid function on diabetes, the control of diabetes, and the prevention and treatment of complications have important clinical significance.
... It has been reported that COVID-19 may increase the risk of developing diabetic ketoacidosis and hyperglycemic hyperosmolar syndrome, even in people without previously diagnosed DM. Along with this, there is a tendency to hypocoagulation protrombotic condition in patients with DM against the background of COVID-19 [54]. Insulin resistance and DM contribute to atherothrombotic conditions as a result of imbalance of factors that regulate coagulation and fibrinolysis. ...
Article
Full-text available
Background. The purpose is to analyze and summarize the studies conducted to determine the impact of COVID-19 on the course of diabetes mellitus and the manifestations of chronic complications of this disease. Materials and methods. We did the search in PubMed and ScienceDirect databases using the following line: “SARS-CoV-2 or COVID-19 and diabetes or hyperglycemia”. Results. It was found that in patients with diabetes, the severity of COVID-19 is increased. In particular, the number of those who needed intensive care or artificial ventilation has elevated. The same negative trend is noted for the overall mortality rate in patients with diabetes. It has been found that COVID-19 contributes to an increase in the development of diabetes, including diabetic ketoacidosis, hyperosmolar syndrome, brain stroke and mental health disorders, which can potentiate the effect of COVID-19 in enhancing the manifestations of diabetes and double risk for these patients. Along with this, the common pathways of the pathogenesis of diabetes and COVID-19 determine the bidirectionality of the interaction in their comorbidity. Conclusions. We have demonstrated numerous links between COVID-19 pathogenetic mechanisms and diabetes. Despite this, current research shows only modest evidence of an increased risk of metabolic, neurological, and psychiatric complications in patients with diabetes who had COVID-19. However, along with the direct impact of patients’ infection, the conditions of pandemic are also affected, which complicate access to the necessary care and should be taken into account when studying the clinical consequences of COVID-19 in patients with diabetes.
Article
This study aims to apply machine learning models to identify new biomarkers associated with the early diagnosis and prognosis of SARS-CoV-2 infection.Plasma and serum samples from COVID-19 patients (mild, moderate, and severe), patients with other pneumonia (but with negative COVID-19 RT-PCR), and healthy volunteers (control) from hospitals in four different countries (China, Spain, France, and Italy) were analyzed by GC–MS, LC–MS, and NMR. Machine learning models (PCA and PLS-DA) were developed to predict the diagnosis and prognosis of COVID-19 and identify biomarkers associated with these outcomes.A total of 1410 patient samples were analyzed. The PLS-DA model presented a diagnostic and prognostic accuracy of around 95% of all analyzed data. A total of 23 biomarkers (e.g., spermidine, taurine, l-aspartic, l-glutamic, l-phenylalanine and xanthine, ornithine, and ribothimidine) have been identified as being associated with the diagnosis and prognosis of COVID-19. Additionally, we also identified for the first time five new biomarkers (N-Acetyl-4-O-acetylneuraminic acid, N-Acetyl-l-Alanine, N-Acetyltriptophan, palmitoylcarnitine, and glycerol 1-myristate) that are also associated with the severity and diagnosis of COVID-19. These five new biomarkers were elevated in severe COVID-19 patients compared to patients with mild disease or healthy volunteers.The PLS-DA model was able to predict the diagnosis and prognosis of COVID-19 around 95%. Additionally, our investigation pinpointed five novel potential biomarkers linked to the diagnosis and prognosis of COVID-19: N-Acetyl-4-O-acetylneuraminic acid, N-Acetyl-l-Alanine, N-Acetyltriptophan, palmitoylcarnitine, and glycerol 1-myristate. These biomarkers exhibited heightened levels in severe COVID-19 patients compared to those with mild COVID-19 or healthy volunteers.
Article
Full-text available
Background and Objectives The mechanisms underlying COVID‐19‐associated pulmonary mucormycosis (CAPM) remain unclear. We use a transcriptomic analysis of the innate immune cells to investigate the host immune and metabolic response pathways in patients with CAPM. Patients and Methods We enrolled subjects with CAPM ( n = 5), pulmonary mucormycosis (PM) without COVID‐19 ( n = 5), COVID‐19 (without mucormycosis, n = 5), healthy controls ( n = 5) without comorbid illness and negative for SARS‐CoV‐2. Peripheral blood samples from cases were collected before initiating antifungal therapy, and neutrophils and monocytes were isolated. RNA sequencing was performed using Illumina HiSeqX from monocytes and neutrophils. Raw reads were aligned with HISAT‐2 pipeline and DESeq2 was used for differential gene expression. Gene ontology (GO) and metabolic pathway analysis were performed using Shiny GO application and R packages (ggplot2, Pathview). Results The derangement of core immune and metabolic responses in CAPM patients was noted. Pattern recognition receptors, dectin‐2, MCL, FcRγ receptors and CLEC‐2, were upregulated, but signalling pathways such as JAK–STAT, IL‐17 and CARD‐9 were downregulated; mTOR and MAP‐kinase signalling were elevated in monocytes from CAPM patients. The complement receptors, NETosis, and pro‐inflammatory responses, such as S100A8/A9, lipocalin and MMP9, were elevated. The major metabolic pathways of glucose metabolism—glycolysis/gluconeogenesis, pentose phosphate pathway, HIF signalling and iron metabolism‐ferroptosis were also upregulated in CAPM. Conclusions We identified significant alterations in the metabolic pathways possibly leading to cellular iron overload and a hyperglycaemic state. Immune responses revealed altered recognition, signalling, effector functions and a pro‐inflammatory state in monocytes and neutrophils from CAPM patients.
Article
Full-text available
Background The majority of the critically ill patients may have critical illness-related corticosteroid insufficiency (CIRCI). The therapeutic effect of dexamethasone may be related to its ability to improve cortical function. Recent study showed that dexamethasone can reduce COVID-19 deaths by up to one third in critically ill patients. The aim of this article is to investigate whether SARS-CoV-2 can attack the adrenal cortex to aggravate the relative adrenal insufficiency. Methods We summarized the clinical features of COVID-19 reported in currently available observational studies. ACE2 and TMPRSS2 expression was examined in human adrenal glands by immunohistochemical staining. We retrospectively analyzed serum cortisol levels in critically ill patients with or without COVID-19. Results High percentage of critically ill patients with SARS-COV-2 infection in the study were treated with vasopressors. ACE2 receptor and TMPRSS2 serine protease were colocalized in adrenocortical cells in zona fasciculata and zona reticularis. We collected plasma cortisol concentrations in nine critically ill patients with COVID-19. The cortisol levels of critically ill patients with COVID-19 were lower than those in non-COVID-19 critically ill group. Six of the nine COVID-19 critically ill patients had random plasma cortisol concentrations below 10 µg/dl, which met the criteria for the diagnosis of CIRCI. Conclusion We demonstrate that ACE2 and TMPRSS2 are colocalized in adrenocortical cells, and that the cortisol levels are lower in critically ill patients with COVID-19 as compared to those of non-COVID-19 critically ill patients. Based on our findings, we recommend measuring plasma cortisol level to guide hormonal therapy.
Article
Full-text available
Since its emergence, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread worldwide, and led to ever-increasing mortality. SARS-CoV-2 infection perturbs the function of the body's vital organs, making patients of all ages susceptible to the disease. Nevertheless, individuals developing critical illness with poor outcomes were mostly the elderly and people with co-morbid conditions, who constituted the vast majority of coronavirus disease 2019 (COVID-19) fatalities. Complications of COVID-19 mostly involve the respiratory, renal and cardiovascular systems, and in severe cases secondary infections leading to pneumonia and acute respiratory distress syndrome, which may precede the death of the patient. Multi-organ failure in individuals with COVID-19 could be a consequence of their co-morbidities. A patient's pre-existing conditions may affect the disease prognosis, requiring immediate attention to accurately detect and evaluate them in SARS-CoV-2-infected individuals. This review addresses several issues in relation to manifestations of the body's vital organs along with potential diagnostic blood factors in SARS-CoV-2 infection. It is hoped that the review will lead to more comprehensive understanding of this complex disease.
Article
Full-text available
Background: Cardiometabolic disorders may worsen Covid-19 outcomes. We investigated features and Covid-19 outcomes for patients with or without diabetes, and with or without cardiometabolic multimorbidity. Methods: We collected and compared data retrospectively from patients hospitalized for Covid-19 with and without diabetes, and with and without cardiometabolic multimorbidity (defined as ≥ two of three risk factors of diabetes, hypertension or dyslipidaemia). Multivariate logistic regression was used to assess the risk of the primary composite outcome (any of mechanical ventilation, admission to an intensive care unit [ICU] or death) in patients with diabetes and in those with cardiometabolic multimorbidity, adjusting for confounders. Results: Of 354 patients enrolled, those with diabetes (n = 81), compared with those without diabetes (n = 273), had characteristics associated with the primary composite outcome that included older age, higher prevalence of hypertension and chronic obstructive pulmonary disease (COPD), higher levels of inflammatory markers and a lower PaO2/FIO2 ratio. The risk of the primary composite outcome in the 277 patients who completed the study as of May 15th, 2020, was higher in those with diabetes (Adjusted Odds Ratio (adjOR) 2.04, 95%CI 1.12-3.73, p = 0.020), hypertension (adjOR 2.31, 95%CI: 1.37-3.92, p = 0.002) and COPD (adjOR 2.67, 95%CI 1.23-5.80, p = 0.013). Patients with cardiometabolic multimorbidity were at higher risk compared to patients with no cardiometabolic conditions (adjOR 3.19 95%CI 1.61-6.34, p = 0.001). The risk for patients with a single cardiometabolic risk factor did not differ with that for patients with no cardiometabolic risk factors (adjOR 1.66, 0.90-3.06, adjp = 0.10). Conclusions: Patients with diabetes hospitalized for Covid-19 present with high-risk features. They are at increased risk of adverse outcomes, likely because diabetes clusters with other cardiometabolic conditions.
Article
Full-text available
Letter to the Editor: Glucose-6-phosphate dehydrogenase deficiency and SARS- CoV-2 mortality: Is there a link and what should we do?
Article
Full-text available
Background: COVID 19-related quarantine led to a sudden and radical lifestyle changes, in particular in eating habits. Objectives of the study were to investigate the effect of quarantine on sleep quality (SQ) and body mass index (BMI), and if change in SQ was related to working modalities. Materials: We enrolled 121 adults (age 44.9 ± 13.3 years and 35.5% males). Anthropometric parameters, working modalities and physical activity were studied. Sleep quality was evaluated by the Pittsburgh Sleep Quality Index (PSQI) questionnaire. At baseline, the enrolled subjects were assessed in outpatient clinic and after 40 days of quarantine/lockdown by phone interview. Results: Overall, 49.6% of the subjects were good sleepers (PSQI < 5) at the baseline and significantly decreased after quarantine (p < 0.001). In detail, sleep onset latency (p < 0.001), sleep efficiency (p = 0.03), sleep disturbances (p < 0.001), and daytime dysfunction (p < 0.001) significantly worsened. There was also a significant increase in BMI values in normal weight (p = 0.023), in subjects grade I (p = 0.027) and II obesity (p = 0.020). In all cohort, physical activity was significantly decreased (p = 0.004). However, analyzing the data according gender difference, males significantly decreased physical activity as well as females in which there was only a trend without reaching statistical significance (53.5% vs 25.6%; p = 0.015 and 50.0% vs 35.9%, p = 0.106; in males and females, respectively). In addition, smart working activity resulted in a significant worsening of SQ, particularly in males (p < 0.001). Conclusions: Quarantine was associated to a worsening of SQ, particularly in males doing smart working, and to an increase in BMI values.
Article
Full-text available
Purpose. Epidemiological data suggest that comorbid patients, mostly those with type 2 diabetes (T2D), are predisposed to poor prognosis in Coronavirus disease 2019 (COVID-19), leading to serious healthcare concerns. The aim of the present manuscript is to review the main relevant mechanisms possibly contributing to worsen the clinical course of COVID-19 in T2D. Results. Poor glucose control, high glycaemic variability and diabetes-related comorbidities at baseline, particularly cardiovascular diseases and obesity, contribute in worsening the prognosis in the above-mentioned cluster of patients. Moreover, both a lower efficient innate immune system response and cytokine dysregulation predispose patients with T2D to impaired viral clearance and more serious pulmonary and systemic inflammation once the SARS-CoV-2 infection occurred. Inconclusive data are currently available for specifically indicate or contraindicate concurrent medications for managing T2D and its comorbidities in infected patients. Conclusions. T2D individuals should be considered as more vulnerable to COVID-19 than general population, and thus require adequate advices about hygienic tips to protect themselves during the pandemic. A careful management of glucose levels and diabetes-related comorbidities remains essential for avoiding further complications, and patient monitoring during the pandemic should be performed also at distance by means of telemedicine. Further studies are needed to clarify whether medications normally used for managing T2D and its associated comorbidities could have a protective or detrimental effect on COVID-19 clinical course.
Article
This article is protected by copyright. All rights reserved. This article is protected by copyright. All rights reserved.
Article
Hyperglycemia with or without blood glucose in diabetes range is an emerging finding not uncommonly encountered in patients with COVID-19. Increasingly, all evidence currently available hints that both new-onset hyperglycemia without diabetes and new-onset diabetes in COVID-19 is associated with a poorer outcome compared with normoglycemic individuals and people with pre-existing diabetes.
Article
Background and Aim Currently, the number of patients with coronavirus disease 2019 (COVID-19) infection is increasing rapidly worldwide. In this study, we aimed to assess whether diabetes mellitus (DM) would increase the risk of severe infection and death in patients with COVID-19. Methods We systematically searched the PubMed, Web of Science, MedRxiv and COVID-19 academic research communication platform for studies reporting clinical severity and/or overall mortality data on DM in patients with COVID-19 published up to July 10, 2020. The primary outcome was to compare the severe infection rate and mortality rate in COVID-19 patients with and without DM, and to calculate the odds ratio (OR) and 95% confidence interval (CI). Results A total of 77 studies involving 32,015 patients with COVID-19 were included in our meta-analysis. COVID-19 patients with DM had higher severe infection and case-mortality rates compared with those without DM (21.4 vs. 10.5% and 28.3 vs. 13.1%, respectively, all p <0.01). COVID-19 patients with DM were at significantly elevated risk of severe infection (OR = 2.49, 95% CI: 2.10-2.95, p <0.001) and mortality (OR = 2.22, 95% CI: 1.80-2.47, p <0.001). Conclusion DM is associated with increased risk of severe infection and higher mortality in patients with COVID-19. Our study suggests that clinicians should pay more attention to the monitoring and treatment of COVID-19 patients with DM.