Abstract
Emerging evidence indicates that the relationship between coronavirus disease 2019 (COVID-19) and diabetes is 2-fold: 1) it is known that the presence of diabetes and other metabolic alterations poses a considerably high risk to develop a severe COVID-19; 2) patients who survived a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection have an increased risk of developing new-onset diabetes. However, the mechanisms underlying this association are mostly unknown, and there are no reliable biomarkers to predict the development of new-onset diabetes. In the present study, we demonstrate that a specific microRNA (miR-34a) contained in circulating extracellular vesicles released by endothelial cells reliably predicts the risk of developing new-onset diabetes in COVID-19. This association was independent of age, sex, body mass index (BMI), hypertension, dyslipidemia, smoking status, and D-dimer.
SIGNIFICANCE STATEMENT
We demonstrate for the first time that a specific microRNA (miR-34a) contained in circulating extracellular vesicles released by endothelial cells is able to reliably predict the risk of developing diabetes after having contracted coronavirus disease 2019 (COVID-19). This association was independent of age, sex, body mass index (BMI), hypertension, dyslipidemia, smoking status, and D-dimer. Our findings are also relevant when considering the emerging importance of post-acute sequelae of COVID-19, with systemic manifestations observed even months after viral negativization (long COVID).
Introduction
The coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to a global healthcare crisis: >110 million Americans and >700 million worldwide have been diagnosed as infected by SARS-CoV-2, leading to ∼1.2 million and ∼7 million deaths, respectively.
The relationship between COVID-19 and diabetes is 2-fold: although it is known that the presence of diabetes and other metabolic alterations poses a considerably high risk to develop a severe COVID-19 (Zhu et al., 2020; Kastora et al., 2022; Kazakou et al., 2022; Kim et al., 2022), emerging evidence indicates that patients who survived a SARS-CoV-2 infection have a significantly increased risk of developing new-onset diabetes (Rubino et al., 2020; Accili, 2021; Kamrath et al., 2021; Khunti et al., 2021; Metwally et al., 2021; Nassar et al., 2021; Sathish et al., 2021; Lai et al., 2022; Rahmati et al., 2022; Rathmann et al., 2022; Ssentongo et al., 2022; Steenblock et al., 2022; Wander et al., 2022; Xie and Al-Aly, 2022; Zhang et al., 2022; Abumayyaleh et al., 2023; Ali et al., 2023; Bellia et al., 2023; Cefalu, 2023; Chourasia et al., 2023; Gorchane et al., 2023; Izzo et al., 2023; Kim et al., 2023; Li et al., 2023; Naveed et al., 2023; O’Mahoney et al., 2022; Weiss et al., 2023; Zhou et al., 2023; Pantea Stoian et al., 2024; Trimarco et al., 2024). Of note, a retrospective analysis of more than 27 million patients (Cerner Real-World Data) revealed that COVID-19 is associated with an increased risk of diabetes and that Black, Asian/Pacific Islander, and American Indian/Alaskan Native populations are disproportionately at risk (Qeadan et al., 2022); a significant rise in new-onset type-2 diabetes has been observed among children and adolescents of Alabama during the COVID-19 pandemic (Schmitt et al., 2022); equally important, a meta-analysis has evidenced that the incidence of diabetic ketoacidosis among pediatric patients has significantly increased during COVID-19 pandemic (Alfayez et al., 2022).
Quantifying the expression levels of microRNAs (miRNAs) shuttled by extracellular vesicles (EVs) has been shown to be extremely valuable in clinical practice (Cha et al., 2021; Esmaeili et al., 2021; Gu et al., 2021; Liao et al., 2021; Liu et al., 2021; Papiewska-Pająk et al., 2021; Silvestro et al., 2021; Sonoda et al., 2021; Wang et al., 2021; Yang and Rhee, 2021; Chen et al., 2022; Pavani et al., 2022; Fullerton et al., 2023). We have recently demonstrated that endothelial EVs (EC-EVs) enriched in miR-24 independently predict cerebrovascular events in COVID-19 (Gambardella et al., 2021). In a preliminary assay comparing circulating levels of EC-EVs of COVID-19 patients who developed diabetes to COVID-19 patients who did not develop diabetes, we identified miR-34a as one of the top upregulated miRNAs. Therefore, we hypothesized the existence of a significant association between the onset of diabetes and plasma levels of EC-EV miR-34a in patients who have been hospitalized for COVID-19.
Materials and Methods
Study Design and Participants.
In this prospective study, we obtained plasma from consecutive patients hospitalized for COVID-19, enrolled at the “Ospedali dei Colli” in Naples, Italy. The diagnosis of diabetes was defined according to the guidelines of the American Diabetes Association (Draznin et al., 2022). The study was performed in accordance with the ethical standards laid down in the 1964 Declaration of Helsinki and its later amendments.
Inclusion criteria: age >18 years old, willingness to participate (informed consent signed by each patient or an authorized representative), and diagnosis of COVID-19 confirmed by reverse-transcription quantitative polymerase chain reaction (RT-qPCR) on nasal swab as described (Fiorentino et al., 2021; Gambardella et al., 2021; Trimarco et al., 2022, 2023).
Exclusion criteria: we excluded patients for whom admission blood samples were unavailable, patients with cancer, patients with a preexistent diagnosis of diabetes or prediabetes (Mone et al., 2023a), patients who were not treated with steroids, and patients who received the first diagnosis of diabetes fewer than 30 days after discontinuation of steroid-based treatments.
Experimental Procedures.
EC-EVs were isolated from plasma via serial centrifugation and magnetic isolation (Miltenyi Biotec, Auburn, CA) as we described and validated (Morelli et al., 2019; Wang et al., 2020; Gambardella et al., 2021), and DNA was extracted as reported earlier (Gambardella et al., 2021; Jankauskas et al., 2023; Mone et al., 2023b); EC-EVs miR-34a levels were quantified via droplet digital polymerase chain reaction (ddPCR) using a QX200 ddPCR System (Bio-Rad, Hercules, CA) following the manufacturer’s instructions. A SARS-CoV-2 test (RT-qPCR) was performed in all subjects to confirm or rule out the COVID-19 diagnosis. Blood levels of interleukin-6 (IL-6), D-dimer, high-sensitivity C-reactive protein (hs-CRP), and tumor necrosis factor α (TNFα) were measured in all patients on admission as we described (Fiorentino et al., 2021; Gambardella et al., 2021).
Statistical Analysis.
All data were analyzed using the SPSS software (version 29.0; SPSS, IBM, Armonk, NY), establishing a significant difference at a P value < 0.05. Data are expressed as means ± S.D. or numbers and percentages. The unpaired two-tailed t test using (when appropriate) Welch’s correction for unequal variances was performed. The χ2 test was applied to compare categorical variables. A multivariable linear regression analysis was used to assess the association between miR-34a and new-onset diabetes, adjusting for potential confounders. Receiver operating characteristic (ROC) curves were analyzed to identify the optimal cutoff value of miR-34a levels, calculating the Youden’s index to integrate sensitivity and specificity information, as we previously described (Santulli et al., 2019; Gambardella et al., 2022).
Results
We obtained plasma from 388 COVID-19 patients. We excluded 73 patients who did not fulfill the aforementioned inclusion/exclusion criteria; consequently, the study was conducted with 315 subjects, with a median follow up of 12 months.
Table 1 reports the main clinical parameters of our population. New onset diabetes was diagnosed in 28 COVID-19 patients. No significant differences in therapeutic management and in the presence of comorbidities were observed. As per our exclusion criteria, all subjects received steroids as standard therapy for hospitalized COVID-19 patients.
TABLE 1.
Main characteristics of our population at hospital admission Data on quantitative parameters are expressed as mean ± standard deviation or as median and interquartile range; data on qualitative characteristics are expressed as absolute numbers and percentage values.
| No New-Onset Diabetes (287) |
New-Onset Diabetes (28) |
P | |
|---|---|---|---|
| Age (years) | 60.7 ± 14.1 | 64.28 ± 15.2 | 0.213 |
| Sex (male, %) | 156 (54.3) | 14 (50) | 0.660 |
| BMI (kg/m2) | 24.8 ± 3.4 | 26.2 ± 3.5 | 0.067 |
| Glycemia (mg/dl) | 100.2 ± 12.1 | 103.1 ± 23.6 | 0.281 |
| SBP (mmHg) | 137.76 ± 19.5 | 142.2 ± 19.3 | 0.114 |
| DBP (mmHg) | 84.46 ± 9.47 | 86.3 ± 14.9 | 0.239 |
| Hypertension (%) | 103 (35.9) | 10 (35.7) | 0.690 |
| Dyslipidemia (%) | 86 (29.9) | 9 (32.1) | 0.811 |
| Current smoking (%) | 126 (43.9) | 12 (42.9) | 0.916 |
| Duration of steroid treatment (days) | 7 (7, 9) | 7 (7, 8) | 0.843 |
| Interval from steroid discontinuation (days) | 166 (137, 193) | 171 (140, 178) | 0.929 |
| D-dimer (μg/ml) | 2.70 ± 1.6 | 2.94 ± 1.8 | 0.463 |
| IL-6 (pg/ml) | 7.5 ± 4.0# | 8.4 ± 5.5# | 0.121 |
| TNFα (pg/ml) | 6.5 ± 4.2 | 5.8 ± 4.7 | 0.271 |
| hs-CRP (μg/ml) | 3.6 ± 3.2# | 4.2 ± 2.9# | 0.144 |
| EC-EV miR-34a (copies/10 nl) | 1867.4 ± 1490 | 3485.8 ± 1998* | 0.001 |
BMI, body mass index; DBP, diastolic blood pressure; EC-EV miR-34a, level of miR-34a within endothelial extracellular vesicles; hs-CRP, high-sensitivity C-reactive protein; IL-6, interleukin-6; SBP, systolic blood pressure; TNFα, tumor necrosis factor α.
*P < 0.05.
We found that circulating levels of EC-EV miR-34a were significantly upregulated (P < 0.001) in patients with versus without new-onset diabetes (Table 1).
Applying ROC curves, we determined that 3300 copies/10 nl was the optimal cutoff value for miR-34a levels to predict new-onset diabetes, yielding the following results: sensitivity 71.43% [95% confidence interval (CI): 51.33% to 86.78%], specificity 98.61% (95% CI: 96.47% to 99.62%), positive predictive value 83.33% (95% CI: 64.76% to 93.15%), and negative predictive value 97.25% (95% CI: 95.17% to 98.45%).
Using a stepwise multiple regression analysis, adjusting for age, sex, body mass index (BMI), hypertension, dyslipidemia, smoking status, and D-dimer (Table 2), the association between EC-EV miR-34a and new-onset diabetes in COVID-19 patients was confirmed (P < 0.001).
TABLE 2.
Multivariable linear regression analysis assessing the association between miR-34a and new-onset diabetes in COVID-19 patients
| B | SE | Beta | t | 95.0% CI for B | P | ||
|---|---|---|---|---|---|---|---|
| Lower Bound | Upper Bound | ||||||
| Age | 0.002 | 0.001 | 0.103 | 1.859 | 0.000 | 0.004 | 0.064 |
| Sex | 0.000 | 0.032 | 0.000 | −0.008 | −0.063 | 0.062 | 0.993 |
| BMI | 0.007 | 0.004 | 0.090 | 1.665 | −0.001 | 0.016 | 0.097 |
| Hypertension | −0.038 | 0.035 | −0.063 | −1.068 | −0.107 | 0.032 | 0.286 |
| Dyslipidemia | 0.017 | 0.036 | 0.027 | 0.472 | −0.054 | 0.088 | 0.637 |
| Smoking | 0.023 | 0.038 | 0.039 | 0.602 | −0.051 | 0.097 | 0.548 |
| D-Dimer | 0.010 | 0.011 | 0.058 | 0.902 | −0.012 | 0.032 | 0.368 |
| EC-EV miR-34a | 5.351 × 10−5 | 0.000 | 0.302 | 5.449 | 0.000 | 0.000 | <0.001 |
BMI, body mass index; EC-EV miR-34a, level of miR-34a within endothelial extracellular vesicles.
Discussion
To the best of our knowledge, this is the first study showing a significant association between EC-EV noncoding RNAs and new-onset diabetes in COVID-19 patients.
In the spring of 2020, we were among the first groups to indicate a link between COVID-19 and endothelial dysfunction (Gambardella et al., 2020; Santulli et al., 2020; Sardu et al., 2020), and our view was later confirmed by other investigators, associating the systemic manifestations of the disease to a direct or indirect involvement of the endothelium (D’Agnillo et al., 2021; Gelzo et al., 2021; Higashikuni et al., 2021; Kalicińska et al., 2021; Qin et al., 2021; Beltrán-Camacho et al., 2022; Casciola-Rosen et al., 2022; Haffke et al., 2022; Kelliher et al., 2022; Ma et al., 2022; Maldonado et al., 2022; Montiel et al., 2022; Otifi and Adiga, 2022; Santoro et al., 2022; Seitz and Ong, 2022; Tarnawski and Ahluwalia, 2022). Indeed, endothelial cells (ECs) express all cofactors necessary for the internalization of SARS-CoV-2 in host cells, including angiotensin-converting enzyme 2 (ACE2), transmembrane protease serine 2, cathepsins B and D, neuropilin-1, vimentin, and others, thereby representing a natural target of SARS-CoV-2 (Gambardella and Santulli, 2021; Mone et al., 2021; Kansakar et al., 2022; Gambardella et al., 2023). Furthermore, the systemic inflammatory viral reaction observed in patients affected by COVID-19 has been shown to be linked to endothelial dysfunction (Bernard et al., 2020; Loo et al., 2021; Sabioni et al., 2021). Hence, COVID-19 affects not only the epithelial cells of the lung parenchyma but also ECs across the whole body, thus leading to a generalized endothelial damage. Such a damage, caused directly by SARS-CoV-2 infection and/or by the ensuing cytokine storm, can shift the vascular equilibrium toward an altered vascular tone and an increased permeabilization; most of these findings have been substantiated from autopsies of COVID-19 patients since the pandemic outbreak (Otifi and Adiga, 2022). Further supporting our theory of a central role of ECs in COVID-19, clinical trials testing whether interventions that ameliorate endothelial dysfunction can have beneficial effects in COVID-19 patients are ongoing and the available results are very encouraging (Adebayo et al., 2021; Fiorentino et al., 2021; Trimarco et al., 2023).
Consistent with our findings, a significantly augmented risk of newly diagnosed diabetes after SARS-CoV-2 infection, independent of steroid use (which was instead linked to transient hyperglycemia), has been recently demonstrated in a large study (Barrett et al., 2022) performed by the Centers for Disease Control and Prevention (CDC). Current COVID-19 guidelines recommend the use of steroids in hospitalized patients up to 10 days or until discharge and should not be prescribed beyond discharge (Huang et al., 2022). Notably, in our study we ruled out cases of transient hyperglycemia due to steroids by including only patients in which the first diagnosis of new-onset diabetes had been formulated at least 1 month after the discontinuation steroid-based therapies.
A Nature paper (Al-Aly et al., 2021) demonstrated that survivors of COVID-19 had a 39% increased likelihood of receiving a new diabetes diagnosis within 6 months after infection compared with noninfected patients (Al-Aly et al., 2021); the cohort included 73,435 COVID-19 patients of the Veterans Health Administration and ∼5 million control subjects who did not have COVID-19 (Al-Aly et al., 2021). The same team of researchers lately published another paper evidencing that people with COVID-19 exhibit an augmented risk of new-onset diabetes that increased according to the severity of the infection as proxied by the care setting: nonhospitalized, hospitalized, and admitted to intensive care (Xie and Al-Aly, 2022). Another study conducted in 47,780 hospitalized patients with COVID-19 found out that these patients had a ∼50% increased likelihood of developing diabetes ∼20 weeks after discharge compared with matched control patients (Ayoubkhani et al., 2021). In full alignment with these reports, we demonstrated in a population of more than 200,000 individuals that the risk of new-onset diabetes is markedly increased when comparing the years of the pandemic (2020–2022) to the triennium 2017–2019 (Izzo et al., 2023). All of these findings provide solid support for a diabetogenic effect of COVID-19 beyond the stress response associated with severe illness.
Our results are particularly relevant if we consider that chronic manifestations of the disease, including metabolic disturbances, have been reported even months after the initial infection occurred (“long COVID”) (Feldman et al., 2020; Iqbal et al., 2021; Montefusco et al., 2021; Raveendran and Misra, 2021; Sathish et al., 2021a; Narayan and Staimez, 2022; Scherer et al., 2022).
Our study is not exempt from limitations, including the relatively small size of our population and brief follow-up; moreover, our findings refer exclusively to hospitalized COVID-19 patients and therefore cannot be generalized to subjects with a mild disease. We also reckon that the exact molecular mechanisms linking EC-EV miR-34a to the development of diabetes need to be defined in dedicated research projects, whereas our study merely identified miR-34a as a novel and independent biomarker of disease; nonetheless, a biomarker-centric approach has been shown to be crucial in drug development (Fader et al., 2021).
In conclusion, we identified a significant association between EC-EV miR-34a and new-onset diabetes, which could be instrumental toward the understanding of the molecular mechanisms underlying the pathophysiology of metabolic complications in COVID-19 as well as long COVID. Further analyses in larger groups are warranted to confirm our results, ratify their prognostic value, and explore the potential role of miR-34a in other metabolic events.
Acknowledgments
The authors thank X. Wang for valuable assistance.
Abbreviations
- CI
confidence interval
- COVID-19
coronavirus disease 2019
- EC
endothelial cell
- EC-EV
endothelial extracellular vesicle
- EV
extracellular vesicle
- SARS-CoV-2
severe acute respiratory syndrome coronavirus 2
Authorship Contributions
Participated in research design: Jankauskas, Gambardella, Santulli.
Conducted experiments: Mone, Jankauskas, Manzi, Gambardella, Coppola, Kansakar, Fiorentino, Lombardi, Varzideh, Sorriento, Trimarco, Santulli.
Contributed new reagents or analytic tools: Mone, Jankauskas, Manzi, Gambardella, Coppola, Kansakar, Fiorentino, Lombardi, Varzideh, Sorriento, Trimarco, Santulli.
Performed data analysis: Izzo, Santulli.
Wrote or contributed to the writing of the manuscript: Mone, Jankauskas, Santulli.
Footnotes
Santulli’s Laboratory is supported in part by National Institutes of Health National Heart, Lung, and Blood Institute [Grant R01-HL159062], [Grant R01-HL146691], and [Grant T32-HL144456] and National Institute of Diabetes and Digestive and Kidney Diseases [Grant R01-DK123259] and [Grant R01-DK033823] (to G.S.), by the Diabetes Action Research and Education Foundation (to G.S.), by the Samuel Waxman Foundation (to G.S.), and by the Monique Weill-Caulier and Irma T. Hirschl Trusts (to G.S.). This publication was also produced with the cofunding of the European Union – Next Generation EU, in the context of The National Recovery and Resilience Plan, PE8 Investment, Project Age-It (Ageing Well in an Ageing Society) (to P.M.). S.S.J., J.G., U.K., and F.V. are supported in part by postdoctoral fellowships of the American Heart Association (AHA-836407, AHA-35211151, AHA-1026190, and AHA-241195524).
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
1P.M., S.S.J., and M.V.M. share the first authorship.
References
- Abumayyaleh MNúñez Gil IJViana-Llamas MCRaposeiras Roubin SRomero RAlfonso-Rodríguez EUribarri AFeltes GBecerra-Muñoz VMSantoro F, et al. ; HOPE COVID-19 investigators (2023) Post-COVID-19 syndrome and diabetes mellitus: a propensity-matched analysis of the International HOPE-II COVID-19 Registry. Front Endocrinol (Lausanne) 14:1167087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Accili D (2021) Can COVID-19 cause diabetes? Nat Metab 3:123–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Adebayo AVarzideh FWilson SGambardella JEacobacci MJankauskas SSDonkor KKansakar UTrimarco VMone P, et al. (2021) L-arginine and COVID-19: an update. Nutrients 13:3951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Al-Aly Z, Xie Y, Bowe B (2021) High-dimensional characterization of post-acute sequelae of COVID-19. Nature 594:259–264. [DOI] [PubMed] [Google Scholar]
- Alfayez OM, Aldmasi KS, Alruwais NH, Bin Awad NM, Al Yami MS, Almohammed OA, Almutairi AR (2022) Incidence of diabetic ketoacidosis among pediatrics with type 1 diabetes prior to and during COVID-19 pandemic: a meta-analysis of observational studies. Front Endocrinol (Lausanne) 13:856958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ali H, Abu-Farha M, Hofmann S (2023) Editorial: COVID-19 and diabetes, volume II. Front Endocrinol (Lausanne) 14:1136997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ayoubkhani D, Khunti K, Nafilyan V, Maddox T, Humberstone B, Diamond I, Banerjee A (2021) Post-covid syndrome in individuals admitted to hospital with covid-19: retrospective cohort study. BMJ 372:n693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barrett CEKoyama AKAlvarez PChow WLundeen EAPerrine CGPavkov MERolka DBWiltz JLBull-Otterson L, et al. (2022) Risk for newly diagnosed diabetes >30 days after SARS-CoV-2 infection among persons aged <18 years - United States, March 1, 2020-June 28, 2021. MMWR Morb Mortal Wkly Rep 71:59–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bellia C, Andreadi A, D’Ippolito I, Scola L, Barraco S, Meloni M, Lauro D, Bellia A (2023) Prevalence and risk of new-onset diabetes mellitus after COVID-19: a systematic review and meta-analysis. Front Endocrinol (Lausanne) 14:1215879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beltrán-Camacho LEslava-Alcón SRojas-Torres MSánchez-Morillo DMartinez-Nicolás MPMartín-Bermejo Vde la Torre IGBerrocoso EMoreno JAMoreno-Luna R, et al. (2022) The serum of COVID-19 asymptomatic patients up-regulates proteins related to endothelial dysfunction and viral response in circulating angiogenic cells ex-vivo. Mol Med 28:40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bernard I, Limonta D, Mahal LK, Hobman TC (2020) Endothelium infection and dysregulation by SARS-CoV-2: evidence and caveats in COVID-19. Viruses 13:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Casciola-Rosen LThiemann DRAndrade FTrejo-Zambrano MILeonard EKSpangler JBSkinner NEBailey JYegnasubramanian SWang R, et al. (2022) IgM anti-ACE2 autoantibodies in severe COVID-19 activate complement and perturb vascular endothelial function. JCI Insight 7:e158362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cefalu WT (2023) COVID-19 and rising incidence of diabetes: despite evolving data, an enigma still to be solved. Diabetes Care 46:913–915. [DOI] [PubMed] [Google Scholar]
- Cha S, Seo EH, Lee SH, Kim KS, Oh CS, Moon JS, Kim JK (2021) MicroRNA expression in extracellular vesicles from nasal lavage fluid in chronic rhinosinusitis. Biomedicines 9:471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen W, Zhang F, Hou X, Xu H, Tang D (2022) Ameliorating role of microRNA-378 carried by umbilical cord mesenchymal stem cells-released extracellular vesicles in mesangial proliferative glomerulonephritis. Cell Commun Signal 20:28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chourasia P, Goyal L, Kansal D, Roy S, Singh R, Mahata I, Sheikh AB, Shekhar R (2023) Risk of new-onset diabetes mellitus as a post-COVID-19 condition and possible mechanisms: a scoping review. J Clin Med 12:1159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- D’Agnillo FWalters KAXiao YSheng ZMScherler KPark JGygli SRosas LASadtler KKalish H, et al. (2021) Lung epithelial and endothelial damage, loss of tissue repair, inhibition of fibrinolysis, and cellular senescence in fatal COVID-19. Sci Transl Med 13:eabj7790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Draznin BAroda VRBakris GBenson GBrown FMFreeman RGreen JHuang EIsaacs DKahan S, et al. ; American Diabetes Association Professional Practice Committee (2022) 2. Classification and diagnosis of diabetes: standards of medical care in diabetes-2022. Diabetes Care 45 (Suppl 1):S17–S38. [DOI] [PubMed] [Google Scholar]
- Esmaeili A, Hosseini S, Baghaban Eslaminejad M (2021) Engineered-extracellular vesicles as an optimistic tool for microRNA delivery for osteoarthritis treatment. Cell Mol Life Sci 78:79–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fader KA, Zhang J, Menetski JP, Thadhani RI, Antman EM, Friedman GS, Ramaiah SK, Vaidya VS (2021) A biomarker-centric approach to drug discovery and development: lessons learned from the coronavirus disease 2019 pandemic. J Pharmacol Exp Ther 376:12–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feldman EL, Savelieff MG, Hayek SS, Pennathur S, Kretzler M, Pop-Busui R (2020) COVID-19 and diabetes: a collision and collusion of two diseases. Diabetes 69:2549–2565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fiorentino G, Coppola A, Izzo R, Annunziata A, Bernardo M, Lombardi A, Trimarco V, Santulli G, Trimarco B (2021) Effects of adding L-arginine orally to standard therapy in patients with COVID-19: A randomized, double-blind, placebo-controlled, parallel-group trial. Results of the first interim analysis. EClinicalMedicine 40:101125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fullerton JL, Cosgrove CC, Rooney RA, Work LM (2023) Extracellular vesicles and their microRNA cargo in ischaemic stroke. J Physiol 601:4907–4921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gambardella J, Coppola A, Izzo R, Fiorentino G, Trimarco B, Santulli G (2021) Role of endothelial miR-24 in COVID-19 cerebrovascular events. Crit Care 25:306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gambardella J, Jankauskas SS, D’Ascia SL, Sardu C, Matarese A, Minicucci F, Mone P, Santulli G (2022) Glycation of ryanodine receptor in circulating lymphocytes predicts the response to cardiac resynchronization therapy. J Heart Lung Transplant 41:438–441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gambardella JKansakar USardu CMessina VJankauskas SSMarfella RMaggi PWang XMone PPaolisso G, et al. (2023) Exosomal miR-145 and miR-885 regulate thrombosis in COVID-19. J Pharmacol Exp Ther 384:109–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gambardella J, Khondkar W, Morelli MB, Wang X, Santulli G, Trimarco V (2020) Arginine and endothelial function. Biomedicines 8:277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gambardella J, Santulli G (2021) What is linking COVID-19 and endothelial dysfunction? Updates on nanomedicine and bioengineering from the 2020 AHA Scientific Sessions. Eur Heart J Cardiovasc Pharmacother 7:e2–e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gelzo MCacciapuoti SPinchera BDe Rosa ACernera GScialò FComegna MMormile MFabbrocini GParrella R, et al. (2021) Further findings concerning endothelial damage in COVID-19 patients. Biomolecules 11:1368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gorchane A, Ach T, Sahli J, Abdelkrim AB, Mallouli M, Bellazreg F, Hachfi W, Chaieb MC, Ach K (2023) Uncovering the alarming rise of diabetic ketoacidosis during COVID-19 pandemic: a pioneer African study and review of literature. Front Endocrinol (Lausanne) 14:1234256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gu C, Zhang H, Gao Y (2021) Adipose mesenchymal stem cells-secreted extracellular vesicles containing microRNA-192 delays diabetic retinopathy by targeting ITGA1. J Cell Physiol 236:5036–5051. [DOI] [PubMed] [Google Scholar]
- Haffke MFreitag HRudolf GSeifert MDoehner WScherbakov NHanitsch LWittke KBauer SKonietschke F, et al. (2022) Endothelial dysfunction and altered endothelial biomarkers in patients with post-COVID-19 syndrome and chronic fatigue syndrome (ME/CFS). J Transl Med 20:138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Higashikuni Y, Liu W, Obana T, Sata M (2021) Pathogenic basis of thromboinflammation and endothelial injury in COVID-19: current findings and therapeutic implications. Int J Mol Sci 22:12081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang CW, Yu AS, Song H, Park JS, Wu SS, Khang VK, Subject CC, Shen E (2022) Association between dexamethasone treatment after hospital discharge for patients with COVID-19 infection and rates of hospital readmission and mortality. JAMA Netw Open 5:e221455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iqbal FM, Lam K, Sounderajah V, Clarke JM, Ashrafian H, Darzi A (2021) Characteristics and predictors of acute and chronic post-COVID syndrome: a systematic review and meta-analysis. EClinicalMedicine 36:100899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Izzo RPacella DTrimarco VManzi MVLombardi APiccinocchi RGallo PEsposito GLembo MPiccinocchi G, et al. (2023) Incidence of type 2 diabetes before and during the COVID-19 pandemic in Naples, Italy: a longitudinal cohort study. EClinicalMedicine 66:102345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jankauskas SSMone PAvvisato RVarzideh FDe Gennaro SSalemme LMacina GKansakar UCioppa AFrullone S, et al. (2023) miR-181c targets Parkin and SMAD7 in human cardiac fibroblasts: validation of differential microRNA expression in patients with diabetes and heart failure with preserved ejection fraction. Mech Ageing Dev 212:111818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalicińska EBiernat MRybka JZińczuk AJanocha-Litwin JRosiek-Biegus MMorawska MWaszczuk-Gajda ADrozd-Sokołowska JSzukalski Ł, et al. (2021) Endothelial activation and stress index (EASIX) as an early predictor for mortality and overall survival in hematological and non-hematological patients with COVID-19: multicenter cohort study. J Clin Med 10:4373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kamrath CRosenbauer JEckert AJPappa AReschke FRohrer TRMonkemoller KWurm MHake KRaile K, et al. (2021) Incidence of COVID-19 and risk of diabetic ketoacidosis in new-onset type 1 diabetes. Pediatrics 148:e2021050856. [DOI] [PubMed] [Google Scholar]
- Kansakar U, Gambardella J, Varzideh F, Avvisato R, Jankauskas SS, Mone P, Matarese A, Santulli G (2022) miR-142 targets TIM-1 in human endothelial cells: potential implications for stroke, COVID-19, Zika, Ebola, dengue, and other viral infections. Int J Mol Sci 23:10242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kastora S, Patel M, Carter B, Delibegovic M, Myint PK (2022) Impact of diabetes on COVID-19 mortality and hospital outcomes from a global perspective: an umbrella systematic review and meta-analysis. Endocrinol Diabetes Metab 5:e00338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kazakou P, Lambadiari V, Ikonomidis I, Kountouri A, Panagopoulos G, Athanasopoulos S, Korompoki E, Kalomenidis I, Dimopoulos MA, Mitrakou A (2022) Diabetes and COVID-19; a bidirectional interplay. Front Endocrinol (Lausanne) 13:780663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kelliher SWeiss LCullivan SO’Rourke EMurphy CAToolan SLennon ÁSzklanna PBComer SPMacleod H, et al. (2022) Non-severe COVID-19 is associated with endothelial damage and hypercoagulability despite pharmacological thromboprophylaxis. J Thromb Haemost 20:1008–1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Khunti K, Del Prato S, Mathieu C, Kahn SE, Gabbay RA, Buse JB (2021) COVID-19, hyperglycemia, and new-onset diabetes. Diabetes Care 44:2645–2655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim D, Alshuwaykh O, Sandhu KK, Dennis BB, Cholankeril G, Ahmed A (2022) Trends in all-cause and cause-specific mortality among individuals with diabetes before and during the COVID-19 pandemic in the U.S. Diabetes Care 45:e107–e109. [DOI] [PubMed] [Google Scholar]
- Kim SH, Arora I, Hsia DS, Knowler WC, LeBlanc E, Mylonakis E, Pratley R, Pittas AG (2023) New-onset diabetes after COVID-19. J Clin Endocrinol Metab 108:e1164–e1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lai HYang MSun MPan BWang QWang JTian JDing GYang KSong X, et al. (2022) Risk of incident diabetes after COVID-19 infection: a systematic review and meta-analysis. Metabolism 137:155330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, Peng L, Gu W (2023) The published trend of studies on COVID-19 and diabetes: bibliometric analysis. Front Endocrinol (Lausanne) 14:1248676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liao TL, Chen YM, Tang KT, Chen PK, Liu HJ, Chen DY (2021) MicroRNA-223 inhibits neutrophil extracellular traps formation through regulating calcium influx and small extracellular vesicles transmission. Sci Rep 11:15676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Y, Li YP, Xiao LM, Chen LK, Zheng SY, Zeng EM, Xu CH (2021) Extracellular vesicles derived from M2 microglia reduce ischemic brain injury through microRNA-135a-5p/TXNIP/NLRP3 axis. Lab Invest 101:837–850. [DOI] [PubMed] [Google Scholar]
- Loo J, Spittle DA, Newnham M (2021) COVID-19, immunothrombosis and venous thromboembolism: biological mechanisms. Thorax 76:412–420. [DOI] [PubMed] [Google Scholar]
- Ma Z, Yang KY, Huang Y, Lui KO (2022) Endothelial contribution to COVID-19: an update on mechanisms and therapeutic implications. J Mol Cell Cardiol 164:69–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maldonado FMorales DDíaz-Papapietro CValdés CFernandez CValls NLazo MEspinoza CGonzález RGutiérrez R, et al. (2022) Relationship between endothelial and angiogenesis biomarkers envisage mortality in a prospective cohort of COVID-19 patients requiring respiratory support. Front Med (Lausanne) 9:826218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Metwally AA, Mehta P, Johnson BS, Nagarjuna A, Snyder MP (2021) COVID-19-induced new-onset diabetes: trends and technologies. Diabetes 70:2733–2744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mone P, De Gennaro S, Moriello D, Frullone S, D’Amelio R, Ferrante MNV, Marro A, Santulli G (2023a) Insulin resistance drives cognitive impairment in hypertensive pre-diabetic frail elders: the CENTENNIAL study. Eur J Prev Cardiol 30:1283–1288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mone P, Gambardella J, Wang X, Jankauskas SS, Matarese A, Santulli G (2021) miR-24 targets the transmembrane glycoprotein neuropilin-1 in human brain microvascular endothelial cells. Noncoding RNA 7:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mone PLombardi AKansakar UVarzideh FJankauskas SSPansini AMarzocco SDe Gennaro SFamiglietti MMacina G, et al. (2023b) Empagliflozin improves the microRNA signature of endothelial dysfunction in patients with heart failure with preserved ejection fraction and diabetes. J Pharmacol Exp Ther 384:116–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Montefusco LBen Nasr MD’Addio FLoretelli CRossi APastore IDaniele GAbdelsalam AMaestroni ADell’Acqua M, et al. (2021) Acute and long-term disruption of glycometabolic control after SARS-CoV-2 infection. Nat Metab 3:774–785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Montiel VLobysheva IGérard LVermeersch MPerez-Morga DCastelein TMesland JBHantson PCollienne CGruson D, et al. (2022) Oxidative stress-induced endothelial dysfunction and decreased vascular nitric oxide in COVID-19 patients. EBioMedicine 77:103893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morelli MB, Shu J, Sardu C, Matarese A, Santulli G (2019) Cardiosomal microRNAs are essential in post-infarction myofibroblast phenoconversion. Int J Mol Sci 21:201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Narayan KMV, Staimez LR (2022) Rising diabetes diagnosis in long COVID. Lancet Diabetes Endocrinol 10:298–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nassar M, Nso N, Baraka B, Alfishawy M, Mohamed M, Nyabera A, Sachmechi I (2021) The association between COVID-19 and type 1 diabetes mellitus: a systematic review. Diabetes Metab Syndr 15:447–454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Naveed Z, Velásquez García HA, Wong S, Wilton J, McKee G, Mahmood B, Binka M, Rasali D, Janjua NZ (2023) Association of COVID-19 infection with incident diabetes. JAMA Netw Open 6:e238866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Mahoney LLRouten AGillies CEkezie WWelford AZhang AKaramchandani USimms-Williams NCassambai SArdavani A, et al. (2022) The prevalence and long-term health effects of long Covid among hospitalised and non-hospitalised populations: a systematic review and meta-analysis. EClinicalMedicine 55:101762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Otifi HM, Adiga BK (2022) Endothelial dysfunction in Covid-19 infection. Am J Med Sci 363:281–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pantea Stoian ABica ICSalmen TAl Mahmeed WAl-Rasadi KAl-Alawi KBanach MBanerjee YCeriello ACesur M, et al. ; Cardiometabolic Panel of International Experts on Syndemic COVID-19 (CAPISCO) (2024) New-onset diabetes mellitus in COVID-19: a scoping review. Diabetes Ther 15:33–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Papiewska-Pająk I, Przygodzka P, Krzyżanowski D, Soboska K, Szulc-Kiełbik I, Stasikowska-Kanicka O, Boncela J, Wągrowska-Danilewicz M, Kowalska MA (2021) Snail overexpression alters the microRNA content of extracellular vesicles released from HT29 colorectal cancer cells and activates pro-inflammatory state in vivo. Cancers (Basel) 13:172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pavani KCMeese TPascottini OBGuan XLin XPeelman LHamacher JVan Nieuwerburgh FDeforce DBoel A, et al. (2022) Hatching is modulated by microRNA-378a-3p derived from extracellular vesicles secreted by blastocysts. Proc Natl Acad Sci USA 119:e2122708119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qeadan F, Tingey B, Egbert J, Pezzolesi MG, Burge MR, Peterson KA, Honda T (2022) The associations between COVID-19 diagnosis, type 1 diabetes, and the risk of diabetic ketoacidosis: a nationwide cohort from the US using the Cerner Real-World Data. PLoS One 17:e0266809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qin ZLiu FBlair RWang CYang HMudd JCurrey JMIwanaga NHe JMi R, et al. (2021) Endothelial cell infection and dysfunction, immune activation in severe COVID-19. Theranostics 11:8076–8091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rahmati M, Keshvari M, Mirnasuri S, Yon DK, Lee SW, Il Shin J, Smith L (2022) The global impact of COVID-19 pandemic on the incidence of pediatric new-onset type 1 diabetes and ketoacidosis: a systematic review and meta-analysis. J Med Virol 94:5112–5127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rathmann W, Kuss O, Kostev K (2022) Incidence of newly diagnosed diabetes after Covid-19. Diabetologia 65:949–954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raveendran AV, Misra A (2021) Post COVID-19 syndrome (“long COVID”) and diabetes: challenges in diagnosis and management. Diabetes Metab Syndr 15:102235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rubino FAmiel SAZimmet PAlberti GBornstein SEckel RHMingrone GBoehm BCooper MEChai Z, et al. (2020) New-onset diabetes in Covid-19. N Engl J Med 383:789–790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sabioni L, De Lorenzo A, Lamas C, Muccillo F, Castro-Faria-Neto HC, Estato V, Tibirica E (2021) Systemic microvascular endothelial dysfunction and disease severity in COVID-19 patients: evaluation by laser Doppler perfusion monitoring and cytokine/chemokine analysis. Microvasc Res 134:104119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santoro LFalsetti LZaccone VNesci ATosato MGiupponi BSavastano MCMoroncini GGasbarrini ALandi F, et al. ; On Behalf Of Gemelli Against Covid-Post-Acute Care Study Group (2022) Impaired endothelial function in convalescent phase of COVID-19: a 3 month follow up observational prospective study. J Clin Med 11:1774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santulli G, Morelli MB, Gambardella J (2020) Is endothelial dysfunction the concealed cornerstone of COVID-19? Rapid response to: Clinical characteristics of 113 deceased patients with coronavirus disease 2019: retrospective study. BMJ 368:m1091. DOI: 10.1136/bmj.m1091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santulli GPascale VFinelli RVisco VGiannotti RMassari AMorisco CCiccarelli MIllario MIaccarino G, et al. (2019) We are what we eat: impact of food from short supply chain on metabolic syndrome. J Clin Med 8:2061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sardu C, Gambardella J, Morelli MB, Wang X, Marfella R, Santulli G (2020) Hypertension, thrombosis, kidney failure, and diabetes: is COVID-19 an endothelial disease? A comprehensive evaluation of clinical and basic evidence. J Clin Med 9:1417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sathish T, Kapoor N, Cao Y, Tapp RJ, Zimmet P (2021) Proportion of newly diagnosed diabetes in COVID-19 patients: a systematic review and meta-analysis. Diabetes Obes Metab 23:870–874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scherer PE, Kirwan JP, Rosen CJ (2022) Post-acute sequelae of COVID-19: a metabolic perspective. eLife 11:e78200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schmitt JA, Ashraf AP, Becker DJ, Sen B (2022) Changes in type 2 diabetes trends in children and adolescents during the COVID-19 pandemic. J Clin Endocrinol Metab 107:e2777–e2782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seitz A, Ong P (2022) Endothelial dysfunction in COVID-19: a potential predictor of long-COVID? Int J Cardiol 349:155–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Silvestro S, Gugliandolo A, Chiricosta L, Diomede F, Trubiani O, Bramanti P, Pizzicannella J, Mazzon E (2021) MicroRNA profiling of HL-1 cardiac cells-derived extracellular vesicles. Cells 10:273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sonoda Y, Kano F, Murata M (2021) Applications of cell resealing to reconstitute microRNA loading to extracellular vesicles. Sci Rep 11:2900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ssentongo P, Zhang Y, Witmer L, Chinchilli VM, Ba DM (2022) Association of COVID-19 with diabetes: a systematic review and meta-analysis. Sci Rep 12:20191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Steenblock CHassanein MKhan EGYaman MKamel MBarbir MLorke DERock JAEverett DBejtullah S, et al. (2022) Diabetes and COVID-19: short- and long-term consequences. Horm Metab Res 54:503–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tarnawski AS, Ahluwalia A (2022) Endothelial cells and blood vessels are major targets for COVID-19-induced tissue injury and spreading to various organs. World J Gastroenterol 28:275–289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trimarco V, Izzo R, Lombardi A, Coppola A, Fiorentino G, Santulli G (2023) Beneficial effects of L-arginine in patients hospitalized for COVID-19: new insights from a randomized clinical trial. Pharmacol Res 191:106702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trimarco VIzzo RPacella DTrama UManzi MVLombardi APiccinocchi RGallo PEsposito GPiccinocchi G, et al. (2024) Incidence of new-onset hypertension before, during, and after the COVID-19 pandemic: a 7-year longitudinal cohort study in a large population. BMC Medicine In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trimarco VIzzo RZanforlin ATursi FScarpelli FSantus PPennisi APelaia GMussi CMininni S, et al. (2022) Endothelial dysfunction in long-COVID: new insights from the nationwide multicenter LINCOLN study. Pharmacol Res 185:106486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wander PL, Lowy E, Beste LA, Tulloch-Palomino L, Korpak A, Peterson AC, Kahn SE, Boyko EJ (2022) The incidence of diabetes among 2,777,768 veterans with and without recent SARS-CoV-2 infection. Diabetes Care 45:782–788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang L, Wang Y, Xiang Y, Ma J, Zhang H, Dai J, Hou Y, Yang Y, Ma J, Li H (2021) An in vitro study on extracellular vesicles from adipose-derived mesenchymal stem cells in protecting stress urinary incontinence through microRNA-93/F3 axis. Front Endocrinol (Lausanne) 12:693977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang X, Morelli MB, Matarese A, Sardu C, Santulli G (2020) Cardiomyocyte-derived exosomal microRNA-92a mediates post-ischemic myofibroblast activation both in vitro and ex vivo. ESC Heart Fail 7:284–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weiss A, Donnachie E, Beyerlein A, Ziegler AG, Bonifacio E (2023) Type 1 diabetes incidence and risk in children with a diagnosis of COVID-19. JAMA 329:2089–2091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xie Y, Al-Aly Z (2022) Risks and burdens of incident diabetes in long COVID: a cohort study. Lancet Diabetes Endocrinol 10:311–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang HC, Rhee WJ (2021) Single step in situ detection of surface protein and microRNA in clustered extracellular vesicles using flow cytometry. J Clin Med 10:319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang T, Mei Q, Zhang Z, Walline JH, Liu Y, Zhu H, Zhang S (2022) Risk for newly diagnosed diabetes after COVID-19: a systematic review and meta-analysis. BMC Med 20:444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou L, Qu H, Zhang Q, Hu J, Shou L (2023) Case report: fulminant type 1 diabetes following paucisymptomatic SARS-CoV-2 infection during late pregnancy. Front Endocrinol (Lausanne) 14:1168927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu LShe ZGCheng XQin JJZhang XJCai JLei FWang HXie JWang W, et al. (2020) Association of blood glucose control and outcomes in patients with COVID-19 and pre-existing type 2 diabetes. Cell Metab 31:1068–1077.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
