Browse

You are looking at 31 - 40 of 87 items for :

  • Metabolic Syndrome and Diabetes x
Clear All
Henry Zelada Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, USA

Search for other papers by Henry Zelada in
Google Scholar
PubMed
Close
,
M Citlalli Perez-Guzman Internal Medicine Division of Endocrinology, Centro Médico ABC, Mexico City, Mexico

Search for other papers by M Citlalli Perez-Guzman in
Google Scholar
PubMed
Close
,
Daniel R Chernavvsky Center for Diabetes Technology, University of Virginia, Charlottesville, Virginia, USA

Search for other papers by Daniel R Chernavvsky in
Google Scholar
PubMed
Close
, and
Rodolfo J Galindo Division of Endocrinology, Diabetes and Metabolism, University of Miami Miller School of Medicine. Miami, Florida, USA

Search for other papers by Rodolfo J Galindo in
Google Scholar
PubMed
Close

Over the last few years, several exciting changes in continuous glucose monitoring (CGM) technology have expanded its use and made CGM the standard of care for patients with type 1 and type 2 diabetes using insulin therapy. Consequently, hospitals started to notice increased use of these devices in their hospitalized patients. Furthermore during the coronavirus disease 2019 (COVID) pandemic, there was a critical need for innovative approaches to glycemic monitoring, and several hospitals started to implement CGM protocols in their daily practice. Subsequently, a plethora of studies have demonstrated the efficacy and safety of CGM use in the hospital, leading to clinical practice guideline recommendations. Several studies have also suggested that CGM has the potential to become the standard of care for some hospitalized patients, overcoming the limitations of current capillary glucose testing. Albeit, there is a need for more studies and particularly regulatory approval. In this review, we provide a historical overview of the evolution of glycemic monitoring in the hospital and review the current evidence, implementation protocols, and guidance for the use of CGM in hospitalized patients.

Open access
Yee-Ming M Cheung Department of Medicine, The University of Melbourne, Austin Health, Melbourne, Australia
Department of Endocrinology, Austin Health, Melbourne, Australia
Division of Endocrinology, Diabetes and Metabolism, Northwell, Great Neck, New York, USA

Search for other papers by Yee-Ming M Cheung in
Google Scholar
PubMed
Close
,
Rudolf Hoermann Department of Medicine, The University of Melbourne, Austin Health, Melbourne, Australia

Search for other papers by Rudolf Hoermann in
Google Scholar
PubMed
Close
,
Karen Van Department of Endocrinology, Austin Health, Melbourne, Australia

Search for other papers by Karen Van in
Google Scholar
PubMed
Close
,
Damian Wu Department of Medicine, The University of Melbourne, Austin Health, Melbourne, Australia

Search for other papers by Damian Wu in
Google Scholar
PubMed
Close
,
Jenny Healy Department of Medicine, The University of Melbourne, Austin Health, Melbourne, Australia

Search for other papers by Jenny Healy in
Google Scholar
PubMed
Close
,
Bella Halim Department of Endocrinology, Austin Health, Melbourne, Australia

Search for other papers by Bella Halim in
Google Scholar
PubMed
Close
,
Manjri Raval Department of Endocrinology, Austin Health, Melbourne, Australia

Search for other papers by Manjri Raval in
Google Scholar
PubMed
Close
,
Maria McGill Department of Radiology, Austin Health, Melbourne, Australia

Search for other papers by Maria McGill in
Google Scholar
PubMed
Close
,
Ali Al-Fiadh Department of Medicine, The University of Melbourne, Austin Health, Melbourne, Australia
Department of Cardiology, Austin Health, Melbourne Australia

Search for other papers by Ali Al-Fiadh in
Google Scholar
PubMed
Close
,
Michael Chao Olivia Newton-John Cancer Research and Wellness Centre, Austin Health, Melbourne, Australia

Search for other papers by Michael Chao in
Google Scholar
PubMed
Close
,
Shane White Olivia Newton-John Cancer Research and Wellness Centre, Austin Health, Melbourne, Australia

Search for other papers by Shane White in
Google Scholar
PubMed
Close
,
Belinda Yeo Olivia Newton-John Cancer Research and Wellness Centre, Austin Health, Melbourne, Australia
Olivia Newton-John Cancer Research Institute, Austin Health, Melbourne, Australia

Search for other papers by Belinda Yeo in
Google Scholar
PubMed
Close
,
Jeffrey D Zajac Department of Medicine, The University of Melbourne, Austin Health, Melbourne, Australia
Department of Endocrinology, Austin Health, Melbourne, Australia

Search for other papers by Jeffrey D Zajac in
Google Scholar
PubMed
Close
, and
Mathis Grossmann Department of Medicine, The University of Melbourne, Austin Health, Melbourne, Australia
Department of Endocrinology, Austin Health, Melbourne, Australia

Search for other papers by Mathis Grossmann in
Google Scholar
PubMed
Close

Purpose

We previously demonstrated that 12 months of aromatase inhibitor (AI) treatment was not associated with a difference in body composition or other markers of cardiometabolic health when compared to controls. Here we report on the pre-planned extension of the study. The pre-specified primary hypothesis was that AI therapy for 24 months would lead to increased visceral adipose tissue (VAT) area when compared to controls.

Methods

We completed a 12-month extension to our prospective 12-month cohort study of 52 women commencing AI treatment (median age 64.5 years) and 52 women with breast pathology not requiring endocrine therapy (63.5 years). Our primary outcome of interest was VAT area. Secondary and exploratory outcomes included other measures of body composition, hepatic steatosis, measures of atherosclerosis and vascular reactivity. Using mixed models and the addition of a fourth time point, we increased the number of study observations by 79 and were able to rigorously determine the treatment effect.

Results

Among study completers (AI = 39, controls = 40), VAT area was comparable between groups over 24 months, the mean-adjusted difference was −1.54 cm2 (95% CI: −14.9; 11.9, P = 0.79). Both groups demonstrated parallel and continuous increases in VAT area over the observation period that did not diverge or change between groups. No statistically significant difference in our secondary and exploratory outcomes was observed between groups.

Conclusions

While these findings provide reassurance that short-to-medium-term exposure to AI therapy is not associated with metabolically adverse changes when compared to controls, risk evolution should be less focussed on the AI-associated effect and more on the general development of cardiovascular risk over time.

Open access
Molly L Tanenbaum Division of Endocrinology, Gerontology, and Metabolism, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA

Search for other papers by Molly L Tanenbaum in
Google Scholar
PubMed
Close
and
Persis V Commissariat Section on Clinical, Behavioral, and Outcomes Research, Joslin Diabetes Center, Boston, Massachusetts, USA

Search for other papers by Persis V Commissariat in
Google Scholar
PubMed
Close

Diabetes technology continues to advance, with more individuals with type 1 diabetes (T1D) adopting insulin pumps, continuous glucose monitoring (CGM), and automated insulin delivery (AID) systems that integrate real-time glucose data with an algorithm to assist with insulin dosing decisions. These technologies are linked with benefits to glycemic outcomes (e.g. increased time in target range), diabetes management behaviors, and quality of life. However, current devices and systems are not without barriers and hassles for the user. The intent of this review is to describe the personal challenges and reactions that users experience when interacting with current diabetes technologies, which can affect their acceptance and motivation to engage with their devices. This review will discuss user experiences and strategies to address three main areas: (i) the emotional burden of utilizing a wearable device; (ii) the perceived and experienced negative social consequences of device use; and (iii) the practical challenges of wearing devices.

Open access
Yang Yu Department of Endocrinology and Metabolism, Huai’an Hospital Affiliated to Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, Jiangsu, China

Search for other papers by Yang Yu in
Google Scholar
PubMed
Close
,
Hairong Hao Department of Endocrinology and Metabolism, Huai’an Hospital Affiliated to Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, Jiangsu, China

Search for other papers by Hairong Hao in
Google Scholar
PubMed
Close
,
Linghui Kong Department of Endocrinology and Metabolism, Huai’an Hospital Affiliated to Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, Jiangsu, China

Search for other papers by Linghui Kong in
Google Scholar
PubMed
Close
,
Jie Zhang Department of Endocrinology and Metabolism, Huai’an Hospital Affiliated to Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, Jiangsu, China

Search for other papers by Jie Zhang in
Google Scholar
PubMed
Close
,
Feng Bai Department of Endocrinology and Metabolism, Huai’an Hospital Affiliated to Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, Jiangsu, China

Search for other papers by Feng Bai in
Google Scholar
PubMed
Close
,
Fei Guo Department of Endocrinology and Metabolism, Huai’an Hospital Affiliated to Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, Jiangsu, China

Search for other papers by Fei Guo in
Google Scholar
PubMed
Close
,
Pan Wei Department of Endocrinology and Metabolism, Huai’an Hospital Affiliated to Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, Jiangsu, China

Search for other papers by Pan Wei in
Google Scholar
PubMed
Close
,
Rui Chen Department of Endocrinology and Metabolism, Huai’an Hospital Affiliated to Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, Jiangsu, China

Search for other papers by Rui Chen in
Google Scholar
PubMed
Close
, and
Wen Hu Department of Endocrinology and Metabolism, Huai’an Hospital Affiliated to Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, Jiangsu, China

Search for other papers by Wen Hu in
Google Scholar
PubMed
Close

Previous studies have shown that the elevated levels of circulating branched-chain amino acids (BCAAs) are associated with the development of insulin resistance and its complications, including obesity, type 2 diabetes, cardiovascular disease and some cancers. However, animal models that can mimic the metabolic state of chronically elevated BCAAs in humans are rare. Therefore, the aim of this study was to establish the above animal model and analyse the metabolic changes associated with high BCAA levels. Sixteen 8-week-old Sprague–Dawley (SD) rats were randomly divided into two groups and given either a high fructose diet or a normal diet. BCAA levels as well as blood glucose and lipid levels were measured at different time points of feeding. The mRNA expression levels of two key enzymes of BCAA catabolism, ACAD (acyl-CoA dehydrogenase) and BCKDH (branched-chain α-keto acid dehydrogenase), were measured by qPCR, and the protein expression levels of these two enzymes were analysed by immunohistochemistry. Finally, the metabolite expression differences between the two groups were analysed by Q300 metabolomics technology. Our study confirms that defects in the catabolic pathways of BCAAs lead to increased levels of circulating BCAAs, resulting in disorders of glucose and lipid metabolism characterized by insulin resistance by affecting metabolic pathways associated with amino acids and bile acids.

Open access
Shaomin Shi Division of Nephrology, The First Affiliated Hospital of Yangtze University, Jingzhou, China

Search for other papers by Shaomin Shi in
Google Scholar
PubMed
Close
,
Xinghua Chen Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China

Search for other papers by Xinghua Chen in
Google Scholar
PubMed
Close
,
Wen Yu Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China

Search for other papers by Wen Yu in
Google Scholar
PubMed
Close
,
Xiaolan Ke Division of Nephrology, The First Affiliated Hospital of Yangtze University, Jingzhou, China

Search for other papers by Xiaolan Ke in
Google Scholar
PubMed
Close
, and
Tean Ma Division of Nephrology, The First Affiliated Hospital of Yangtze University, Jingzhou, China

Search for other papers by Tean Ma in
Google Scholar
PubMed
Close

Protection of podocytes is one of the important means to delay the progression of diabetic nephropathy (DN), and glucagon-like peptide-1 (GLP-1) has been shown to have a protective effect on the kidney in DN models, but whether it has a protective effect on podocytes and the potential mechanisms of action remain largely unknown. In the present study, we established a type 2 diabetes mellitus (T2DM) mouse model by high-fat diet feeding combined with streptozotocin (STZ) induction and administered the intervention for 14 weeks. We found that liraglutide significantly ameliorated podocyte injury in DN mice. Mechanistically, we detected glucagon-like peptide-1 receptor (GLP-1R) protein expression levels in kidney tissues by immunohistochemical staining, immunofluorescence staining, and western blotting and found that podocytes could express GLP-1R and liraglutide treatment could restore GLP-1R expression in the kidney tissues of DN mice. Furthermore, we found that NLRP3-induced inflammation and pyroptosis were positively correlated with podocyte injury in DN mice, and liraglutide inhibited the expression of NLRP3-induced inflammation and pyroptosis-related proteins. Our results suggest that liraglutide protects DN mouse podocytes by regulating GLP-1R in renal tissues and by regulating NLRP3-induced inflammation and pyroptosis.

Open access
Zhaoxiang Liu Department of Endocrinology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China

Search for other papers by Zhaoxiang Liu in
Google Scholar
PubMed
Close
,
Mingqiang Zhang Department of Respiratory and Critical Care Medicine, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China

Search for other papers by Mingqiang Zhang in
Google Scholar
PubMed
Close
,
Xiaohu Shi Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China

Search for other papers by Xiaohu Shi in
Google Scholar
PubMed
Close
,
Wenhui Zhao Department of Endocrinology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China

Search for other papers by Wenhui Zhao in
Google Scholar
PubMed
Close
,
Chenxiang Cao Department of Endocrinology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China

Search for other papers by Chenxiang Cao in
Google Scholar
PubMed
Close
,
Lixia Jin Department of Endocrinology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China

Search for other papers by Lixia Jin in
Google Scholar
PubMed
Close
,
Yanlei Wang Department of Endocrinology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China

Search for other papers by Yanlei Wang in
Google Scholar
PubMed
Close
, and
Jianzhong Xiao Department of Endocrinology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China

Search for other papers by Jianzhong Xiao in
Google Scholar
PubMed
Close

Objectives

The activation of immune cells plays a significant role in the progression of type 2 diabetes. This study aimed to investigate the potential role of myeloid-derived suppressor cells (MDSCs) and T-regulatory cells (Tregs) in type 2 diabetes.

Methods

A total of 61 patients diagnosed with type 2 diabetes were recruited. Clinical characteristics were reviewed and peripheral blood samples were collected. We calculated the percentage of different cells. Frequencies of MDSC subsets refered to the percentage of G-MDSCs (CD15+CD33+CD11b+CD14-HLA-DR-/low) in CD45 positive cells and the percentage of M-MDSCs (CD14+CD15-CD11b+CD33+HLA-DR-/low) in lymphocytes plus monocytes.

Results

Frequencies of programmed cell death ligand 1-positive granulocytic MDSCs (PD-L1+ G-MDSCs), programmed cell death ligand 2-positive monocytic MDSCs (PD-L2+ M-MDSCs), PD-L2+ G-MDSC, and programmed cell death protein 1-positive Tregs (PD-1+Tregs) were decreased in patients with type 2 diabetes. The frequency of PD-1+ Tregs was positively related to PD-L2+ M-MDSCs (r= 0.357, P = 0.009) and negatively related to HbA1c (r = -0.265, P = 0.042), fasting insulin level (r = −0.260, P = 0.047), and waist circumference (r = −0.373, P = 0.005).

Conclusions

Decreased PD-L2+ M-MDSCs and PD-1+ Tregs may promote effector T cell activation, leading to chronic low-grade inflammation in type 2 diabetes. These findings highlight the contribution of MDSCs and Tregs to the immunopathogenesis of type 2 diabetes and suggest their potential as targets for new therapeutic approaches.

Open access
Ying Pei Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China

Search for other papers by Ying Pei in
Google Scholar
PubMed
Close
,
Rui Wang School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China

Search for other papers by Rui Wang in
Google Scholar
PubMed
Close
,
Wanyu Chen Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China

Search for other papers by Wanyu Chen in
Google Scholar
PubMed
Close
,
Shulin Yi Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China

Search for other papers by Shulin Yi in
Google Scholar
PubMed
Close
,
Chen Huang Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China

Search for other papers by Chen Huang in
Google Scholar
PubMed
Close
,
Shaochan Liang School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China

Search for other papers by Shaochan Liang in
Google Scholar
PubMed
Close
,
Hongying Cao School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China

Search for other papers by Hongying Cao in
Google Scholar
PubMed
Close
,
Yifei Xu Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China

Search for other papers by Yifei Xu in
Google Scholar
PubMed
Close
, and
Bo Tan Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China

Search for other papers by Bo Tan in
Google Scholar
PubMed
Close

Background

Similar to the high-fat diet (HFD), the high-glycemic diet (HGD) contributes to the development and progression of type 2 diabetes mellitus (T2DM). However, the effect of HGD on gastrointestinal motility in T2DM and its underlying mechanisms remain unclear.

Methods

Thirty C57BL/6J mice were randomly designated into the normal-feeding diet (NFD) group, HFD group, and HGD group. The plasma glucose, plasma insulin, and gastrointestinal motility were examined. Meanwhile, the tension of isolated colonic smooth muscle rings was calculated, and the gut microbiota was analyzed by 16s rDNA high-throughput sequencing.

Result

After 16 weeks of HGD feeding, obesity, hyperglycemia, insulin resistance, and constipation were observed in HGD mice. Autonomic contraction frequency of the colonic neuromuscular system and electrical field stimulation-induced contractions were reduced in HGD mice. On the contrary, neuronal nitric oxide synthase activity and neuromuscular relaxation were found to be enhanced. Finally, gut microbiota analysis revealed that Rhodospirillaceae abundance significantly increased at the family level in HGD mice. At the genus level, the abundance of Insolitispirillum increased remarkably, whereas Turicibacter abundance decreased significantly in HGD mice.

Conclusion

HGD induced constipation in obese diabetic mice, which we speculated that it may be related to neuromuscular dysmotility and intestinal microbiota dysbiosis.

Open access
Laura Hasse Department of Pediatric Dermatology and Allergology, Children’s Hospital Auf der Bult, Hannover, Germany

Search for other papers by Laura Hasse in
Google Scholar
PubMed
Close
,
Dagmar Jamiolkowski Department of Pediatric Dermatology and Allergology, Children’s Hospital Auf der Bult, Hannover, Germany

Search for other papers by Dagmar Jamiolkowski in
Google Scholar
PubMed
Close
,
Felix Reschke Department of Pediatric Diabetology, Children’s Hospital Auf der Bult, Hannover, Germany

Search for other papers by Felix Reschke in
Google Scholar
PubMed
Close
,
Kerstin Kapitzke Department of Pediatric Diabetology, Children’s Hospital Auf der Bult, Hannover, Germany

Search for other papers by Kerstin Kapitzke in
Google Scholar
PubMed
Close
,
Jantje Weiskorn Department of Pediatric Diabetology, Children’s Hospital Auf der Bult, Hannover, Germany

Search for other papers by Jantje Weiskorn in
Google Scholar
PubMed
Close
,
Olga Kordonouri Department of Pediatric Diabetology, Children’s Hospital Auf der Bult, Hannover, Germany

Search for other papers by Olga Kordonouri in
Google Scholar
PubMed
Close
,
Torben Biester Department of Pediatric Diabetology, Children’s Hospital Auf der Bult, Hannover, Germany

Search for other papers by Torben Biester in
Google Scholar
PubMed
Close
, and
Hagen Ott Department of Pediatric Dermatology and Allergology, Children’s Hospital Auf der Bult, Hannover, Germany

Search for other papers by Hagen Ott in
Google Scholar
PubMed
Close

Objective

Little is known about specific cutaneous findings in children and adolescents with overweight and obesity. This study assessed the association of skin signs with pivotal auxological and endocrinological parameters and their influence on the quality of life (QoL) of young people with obesity.

Study design

All patients initially recruited for a tertiary hospital's weight control program were offered participation in this interdisciplinary, single-center, cross-sectional study. All participants underwent a detailed dermatological examination, anthropometric measurements and laboratory examinations. QoL was assessed with validated questionnaires.

Results

A total of 103 children and adolescents (age 11.6 ±2.5 years, 41% female, 25% prepubertal, BMI SDS 2.6 ± 0.5, homeostatic model assessment (HOMA) score 3.3 ± 4.2; mean ± s.d.) were recruited in a 12-month study period. Skin affections were linearly associated with increasing BMI and higher age. The most common skin findings were (%) striae distensae (71.0), keratosis pilaris (64.7), acanthosis nigricans (45.0), acne vulgaris (39.2), acrochordons (25.5) and plantar hyperkeratosis (17.6). The HOMA score was associated with acanthosis nigricans (P = 0.047), keratosis pilaris (P = 0.019) and acne vulgaris (P < 0.001). The general mean QoL(QoL) score, as assessed by the WHO-5, was 70 out of 100. A total of 38.9% of participants reported impaired dermatological QoL.

Conclusions

This study shows the high prevalence of skin lesions in children and adolescents with obesity. The association between skin lesions and the HOMA score indicates that skin manifestations are a marker of insulin resistance. To prevent secondary diseases and improve QoL, thorough skin examinations and interdisciplinary cooperation are necessary.

Open access
Rama Lakshman Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK

Search for other papers by Rama Lakshman in
Google Scholar
PubMed
Close
,
Charlotte Boughton Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
Cambridge University Hospitals NHS Foundation Trust, Wolfson Diabetes and Endocrine Clinic, Cambridge, UK

Search for other papers by Charlotte Boughton in
Google Scholar
PubMed
Close
, and
Roman Hovorka Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK

Search for other papers by Roman Hovorka in
Google Scholar
PubMed
Close

Automated insulin delivery systems, also known as closed-loop or ‘artificial pancreas’ systems, are transforming the management of type 1 diabetes. These systems consist of an algorithm which responds to real-time glucose sensor levels by automatically modulating insulin delivery through an insulin pump. We review the rapidly changing landscape of automated insulin-delivery systems over recent decades, from initial prototypes to the different hybrid closed-loop systems commercially available today. We discuss the growing body of clinical trials and real-world evidence demonstrating their glycaemic and psychosocial benefits. We also address future directions in automated insulin delivery such as dual-hormone systems and adjunct therapy as well as the challenges around ensuring equitable access to closed-loop technology.

Open access
Merlin C Thomas Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia

Search for other papers by Merlin C Thomas in
Google Scholar
PubMed
Close
,
Brendon L Neuen The George Institute for Global Health, Sydney, NSW, Australia

Search for other papers by Brendon L Neuen in
Google Scholar
PubMed
Close
,
Stephen M Twigg The University of Sydney School of Medicine, Sydney, NSW, Australia
Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia

Search for other papers by Stephen M Twigg in
Google Scholar
PubMed
Close
,
Mark E Cooper Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia

Search for other papers by Mark E Cooper in
Google Scholar
PubMed
Close
, and
Sunil V Badve The George Institute for Global Health, Sydney, NSW, Australia
Department of Renal Medicine, St George Hospital, Sydney, NSW, Australia
Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia

Search for other papers by Sunil V Badve in
Google Scholar
PubMed
Close

Sodium‐glucose co-transporter 2 (SGLT2) inhibitors have recently emerged as an effective means to protect kidney function in people with type 2 diabetes and chronic kidney disease (CKD). In this review, we explore the role of SGLT2 inhibition in these individuals. SGLT2 inhibitors specifically act to inhibit sodium and glucose reabsorption in the early proximal tubule of the renal nephron. Although originally developed as glucose-lowering agents through their ability to induce glycosuria, it became apparent in cardiovascular outcome trials that the trajectory of kidney function decline was significantly slowed and the incidence of serious falls in kidney function was reduced in participants receiving an SGLT2 inhibitor. These observations have recently led to specific outcome trials in participants with CKD, including DAPA-CKD, CREDENCE and EMPA-KIDNEY, and real-world studies, like CVD-REAL-3, that have confirmed the observation of kidney benefits in this setting. In response, recent KDIGO Guidelines have recommended the use of SGLT2 inhibitors as first-line therapy in patients with CKD, alongside statins, renin–angiotensin–aldosterone system inhibitors and multifactorial risk factor management as indicated. However, SGLT2 inhibitors remain significantly underutilized in the setting of CKD. Indeed, an inertia paradox exists, with patients with more severe disease less likely to receive an SGLT2 inhibitor. Concerns regarding safety appear unfounded, as acute kidney injury, hyperkalaemia, major acute cardiovascular events and cardiac death in patients with CKD appear to be lower following SGLT2 inhibition. The first-in-class indication of dapagliflozin for CKD may begin a new approach to managing kidney disease in type 2 diabetes.

Open access