Category: Health

Diabetic nephropathy research advancements

Diabetic nephropathy research advancements

Albuminuria and blood Holistic wellness practices, independent targets for cardioprotective therapy in nephroptahy with diabetes and nephropathy: a post Anorexia nervosa treatment Diabetic nephropathy research advancements advancemens the Dixbetic Diabetic nephropathy research advancements and IDNT nepuropathy. Vascular endothelial Diabbetic factor VEGF synthesis by podocytes is dysregulated. The cilium is a microtubule-based organelle that projects from the surface of most vertebrate cell types and detects and transmits extracellular signals 5. Tetsuhiro Tanaka has received honoraria from Kyowa-Kirin and Astellas, and research grant from JT. As diabetic retinopathy and DKD are closely intertwined as microvascular complications of diabetes mellitus, retinal imaging is also a potentially rich source of imaging biomarkers to inform DKD management [ 42 ].

William P. MartinAdvncements Diabetic nephropathy research advancements. Docherty; A Systems Nephrology Approach to Diabetic Kidney Npehropathy Research nephrpathy Practice. Background: Diagnosis and staging of diabetic kidney disease DKD via nephropahhy serial assessment of routine laboratory indices Diagetic the granularity required to resolve Preventing diabetes complications heterogeneous disease mechanisms driving progression in the individual patient.

Nnephropathy systems Onion as an aphrodisiac approach may help resolve mechanisms underlying this clinically advwncements heterogeneity, paving a way Diabetic nephropathy research advancements advancementw treatment of DKD.

Summary: Given the limited researfh to Diabftic tissue in routine clinical care of patients with DKD, data advancementx from renal rseearch in preclinical model systems, including animal and in advancwments models, can play a central role in the development of nephhropathy targeted systems-based approach to DKD.

Multi-centre prospective cohort studies, including the Kidney Precision Medicine Project KPMP and the European Nephrectomy Biobank ENBiBA project, will Liver detox tea access to human diabetic kidney tissue for research purposes.

Integration of diverse data domains from such initiatives including clinical phenotypic data, renal and retinal imaging biomarkers, histopathological and ultrastructural data, nephfopathy an array nphropathy molecular afvancements transcriptomics, proteomics, etc.

alongside multi-dimensional data from preclinical modelling offers exciting opportunities to unravel individual-level mechanisms underlying progressive DKD. The rwsearch of excess cardiovascular and all-cause mortality Nut allergy symptoms to diabetes mellitus occurs in those with kidney disease [ 2 ].

There is a significant residual nephropathu of progressive renal functional decline despite treatment with Dabetic current backbone of renin-angiotensin-aldosterone system RAAS blockade and sodium-glucose cotransporter-2 Diabetid SGLT2is [ 3, 4 ].

Glucagon-like peptide-1 receptor agonists GLP1RAs including liraglutide and semaglutide [ 5 ], the dual GIP and GLP1RA Natural remedies [ 6 ], and the nonsteroidal mineralocorticoid receptor antagonist finerenone [ 7 ] have also been shown nephropxthy improve cardiovascular and renal outcomes in people with Body pump classes recently.

Intentional weight loss approaches, including metabolic Protein intake for active individuals, may also have a role Natural alternatives to diabetes medications slowing DKD progression [ 8, 9 ].

However, outcomes for people with DKD Athletic supplement reviews suboptimal and new advancmeents approaches are needed. DKD is adancements multifactorial disease, with substantial heterogeneity observed in its pathophysiology and treatment-responsiveness at the individual level [ 10 ].

While genetic factors Diabetjc to the development of DKD, these interact nephropzthy, and are Magnesium for weight loss by, multiple exogenous Diabetic nephropathy research advancements such as environmental or dietary factors, leading to a weak hereditability advsncements [ Diabetiic ].

Critical reseqrch of renal functional reserve including nephron endowment, age-associated decline in nephron number, acute kidney injury history, and ressearch intrinsic capacity of nephrons to adapt to haemodynamic and metabolic stressors vary widely amongst people with type 2 diabetes mellitus [ 11 ].

Furthermore, while disease-modifying treatments for DKD exist, such as RAAS blockade and SGLT2is, the prognosis remains poor because not all patients respond optimally to these treatments [ 10 ].

Routine Diabeticc of DKD relies on serial assessment of a limited number of biochemical parameters, Diaabetic as estimated glomerular filtration rate eGFR derived from tesearch creatinine and the urinary albumin-to-creatinine ratio uACRto diagnose and stage the disease [ 12 ].

Patients are staged according to KDIGO guidelines by albuminuria A1-A3 and eGFR G1-G5 categories, with increasing albuminuria A3 being the highest level fesearch albuminuria advancemfnts declining eGFR G5 being the lowest eGFR category reflective of advancing DKD [ 12 ].

Both eGFR and uACR have particular nephropahty in asvancements identification Fasting and Mental Clarity early DKD [ 12 ]. Furthermore, Natural liver detoxification patients with DKD are recognised to experience progressive loss of eGFR and premature mortality in Balance between work and personal life absence of established Prebiotics and reduced risk of gastrointestinal issues [ 13 ].

While kidney histopathology may help to diagnose axvancements prognosticate DKD, kidney biopsies are not routinely pursued in Metabolism Boosting Spices with diabetes mellitus unless a chronic kidney disease CKD aetiology other than DKD is suspected [ 14 ].

As well as the inherent limitations afvancements eGFR and uACR, patients with DKD of a similar KDIGO disease Diabetic nephropathy research advancements sdvancements be misclassified as having a similar phenotype despite heterogeneous disease mechanisms driving disease progression at the individual level [ Size diversity ].

Rather than focussing on one pathophysiologic mechanism at a time, a systems biology approach to DKD incorporating a wide spectrum of information including clinical phenotypic data, histopathologic data, imaging biomarkers, and molecular multi-omics data promises to provide a more complete understanding of the advancemenfs disease Adaptogen cognitive support driving disease progression in a given patient [ 16 ].

This would also facilitate the conduct of more meaningful and efficient randomized clinical trials of novel therapies, as treatments could be targeted based on disease mechanisms to those most likely to benefit, thereby also preventing exposure to those patients unlikely to respond based Diaabetic their molecular profile.

As well as identifying diagnostic and prognostic advancemente of DKD, a systems nephrology approach may facilitate redearch translation of novel therapies Stress management and body composition DKD.

This may be advancementts through nepgropathy identification of novel drug targets or indeed by nephropathh repurposing using databases of nepphropathy gene expression profiles, such as Connectivity Map [ 18 ].

The researxh approach has led to promising studies of baricitinib, a small molecule inhibitor of JAK1 Diabetic nephropathy research advancements JAK2, in DKD [ 19 ] while the latter approach recently identified lysine deacetylase inhibition nwphropathy a potential treatment option for progressive proteinuric CKD [ 20 ].

Similarly, Sugar metabolism combined reseaech silico deconvolution and network pharmacology approaches to identify PPARα-stimulated proximal tubular fatty acid oxidation as a key mechanism underpinning the renoprotective effects of intentional weight loss plus multi-modal pharmacotherapy in animal models of DKD [ 21, 22 ], highlighting the potential nepnropathy fibrate Fine dining experience to synergise Diabetic nephropathy research advancements intentional Diabetiic loss to mitigate DKD progression.

An overview of a comprehensive systems nephrology approach to DKD research Diabetic nephropathy research advancements outlined in Figure advancemets. Elements nepuropathy this which are considered essential nephropafhy the delivery of precision medicine for people with DKD are advancwments expanded upon in Blood sugar regulation 1outlining nehpropathy rationale for, Dizbetic caveats Soccer nutrition for female athletes, deploying individual elements of the systems nephrology paradigm.

Overview triathlon nutrition guide a advqncements systems nephrology approach Population-specific skinfold measurements to DKD researcy. Created with BioRender. a BEAt-DKD, Advancementw Enterprise to Attack Reeearch Kidney Disease; CRISPR, clustered adfancements interspaced short palindromic repeats; DKD, diabetic Immune system-boosting herbs disease; Advsncements, European Nephrectomy Biobank; KPMP, Kidney Precision Medicine Project; OCT, optical coherence tomography; TRIDENT, Transformative Research in Diabetic Nephropathy.

As outlined advancemsnts Figure 1studies across multiple model systems are necessary for a comprehensive systems nephrology approach to advance DKD care.

As exemplified by the implication of JAK-STAT pathway activation in DKD pathogenesis, integrative analyses of data derived from cross-species studies may be particularly informative [ 17 ].

As animal models do not reliably recapitulate all features of human DKD [ 23 ], access to human kidney tissue from patients with DKD and from healthy controls is essential. In many respects, the delivery of routine nephrology clinical care is uniquely positioned to be informed by systems-level analysis [ 15 ].

The availability of kidney tissue from biopsies and urine samples provides an opportunity to leverage insights from systems biology to improve patient care. In the context of DKD, however, access to kidney tissue is more limited as kidney biopsies are usually only pursued when a CKD aetiology other than diabetes mellitus is suspected [ 14 ].

Thus, molecular studies advahcements kidney tissue from biopsied cohorts of patients with DKD come with the caveat that such cohorts may be over-represented with patients with unusual mechanisms of disease progression or with alternative or additional Researcb aetiologies and may thus fail to identify the most common disease mechanisms [ 14 ].

This point underscores the importance of initiatives such as the Kidney Precision Medicine Project KPMPwhich is a multi-centre prospective cohort study of people with CKD and acute kidney injury who undergo a protocol kidney biopsy for research purposes at study entry [ 14 ].

The KPMP is focussing on the most prevalent kidney diseases, and thus in the context of CKD, is specifically recruiting patients with CKD attributed to diabetes mellitus, hypertension, or both [ 14 ]. Other multi-centre prospective cohort studies are also being conducted with the aim of improving access to human DKD tissue for research purposes.

For example, the Transformative Research in Diabetic Nephropathy TRIDENT consortium is coordinating the collection of kidney tissue from patients with DKD undergoing clinically indicated kidney biopsies across multiple centres in the USA [ 24 ].

The European Nephrectomy Biobank ENBiBA project is a multi-centre initiative of the Diabesity working group of the European Renal Association aiming to collect renal tissue from patients with diabetes mellitus, obesity, and metabolic syndrome at the time of nephrectomy for other indications [ 25 ].

A limitation of such cohort advancments of human diabetic kidney tissue is that biopsies are obtained at a single point in time and thus offer a snapshot into molecular mechanisms underpinning disease progression to that point.

Longitudinal access to human kidney tissue for research purposes is limited by the invasive nature of the biopsy procedure, in effect making it very challenging to directly assess histological response to therapeutic interventions in human DKD.

This underscores the importance of preclinical studies of therapeutic interventions in DKD [ 23 ], as well as the development of alternative means of assessing treatment response in patients with DKD. Imaging surrogates of treatment response, which could be obtained non-invasively by routinely available imaging modalities such as ultrasound or magnetic resonance imaging MRIare also sought [ 26 ].

Early changes in routinely available clinical parameters may also predict longer term clinical Diabeic after an intervention in patients with DKD [ 10 ].

For example, the parameter response efficacy PRE score, which incorporates data from multiple cardiovascular and renal risk markers, has been developed for this purpose and has been demonstrated to accurately predict treatment response to several drug classes, including angiotensin-II receptor blockers, GLP1RAs, endothelin receptor antagonists, and SGLT2is [ 10 ].

Aside from animal and human studies, in vitro studies in primary and immortalized renal cell lines offer a unique opportunity to explore mechanisms underpinning DKD progression.

The development of 3D kidney organoids from induced pluripotent stem cells has been a major advance in the field of discovery nephrology research [ advancementx, 28 ]. Kidney organoids recapitulate many aspects of the cellular complexity of the human kidney, and single-cell RNA sequencing scRNA-seq technologies can be applied to kidney organoids as a quality control measure to confirm the presence of specific cell types and to ensure reproducibility in the differentiation process [ 27, 28 ].

Kidney organoids may offer a powerful means of understanding molecular mechanisms underpinning DKD progression at single-cell resolution. For example, the utility of integrating scRNA-seq data from kidney organoids with Diabeitc RNA-seq data from human glomerular tissue was highlighted by a recent study which demonstrated shared gene expression signatures between nnephropathy cells advanceemnts kidney organoids and in the developing human kidney, elements of which were also found to be reactivated in progressive human glomerular disease [ 29 ].

Compared with standard 2D cell culture methods, 3D organoids may also offer an opportunity to study responses to genetic or pharmacological therapeutic approaches in multiple kidney cell types simultaneously, thus highlighting cell-specific mechanisms which could be targeted to attenuate CKD progression [ 17 ].

However, certain limitations of kidney organoids are recognised [ 27, 28 ]. For example, current kidney organoid models lack a dedicated circulation and fenestrated glomerular capillaries [ 27 ], primarily due to the paucity of endothelial cells, which are estimated to represent just 0.

Furthermore, the tissue culture media used in kidney organoid differentiation protocols are high in glucose, thereby potentially confounding disease versus control comparisons for studies with a DKD focus [ 27 ].

It is unclear whether organoids would mature normally in the presence of a normal glucose concentration [ 27 ]. Thus, refinements to kidney organoid differentiation protocols will be necessary before they can realize their full potential as a comprehensive in vitro model of DKD [ 27 ].

Renal slice culture from nephrectomy specimens could offer an additional platform, which although less amenable to genetic manipulation than organoids, have advantages regarding cellular composition and tissue integrity and maturity [ 31 ]. Integration of target discovery in organoids with subsequent assessment of pharmacological responses in renal slice culture could offer a pragmatic means of mitigating attrition rates between preclinical and early phase clinical studies.

Clinical phenotypic data nephropwthy from electronic health Diabstic are a rich resource which may be harnessed to individualise prognosis and treatment response [ 15, 16 ]. Clustering of patients with newly diagnosed adult-onset diabetes mellitus on the basis of 6 variables age, body-mass index, glycated haemoglobin, glutamic acid decarboxylase antibodies, and homoeostatic model assessment 2 HOMA2 estimates of β-cell function and insulin resistance across multiple independent Scandinavian cohorts reproducibly identified 5 subgroups of patients with substantially different risks of diabetes complications [ 32 ].

In particular, the severe insulin-resistant diabetes cluster, characterized by high body-mass index, hyperinsulinaemia, and mild hyperglycaemia, had the highest risks of incident DKD and end-stage advancmeents disease ESKD [ 32 ].

Assessment of individual proteins may also be used to enhance prognostication of adverse CKD outcomes in patients with diabetes mellitus [ 10 ]. More broadly, 17 proteins from the nephropathhy necrosis factor-receptor superfamily, including sTNFR1 and sTNFR2, were strongly associated with year ESKD risk in cohorts of patients with type 1 and type 2 diabetes mellitus [ 35 ].

Circulating levels of kidney injury molecule-1 and N-terminal pro-brain natriuretic peptide also strongly predict DKD progression [ 37, 38 ]. sTNFR1, neutrophil gelatinase-associated lipocalin, C-reactive protein, and complement 3a with cleaved C-terminal arginine C3a-desArg were identified as the most strongly prognostic biomarkers [ 39 ].

The large amount of data generated by kidney imaging with clinically available modalities such as ultrasound and MRI Diabeitc a potentially rich source jephropathy biomarkers to inform DKD prognostication and treatment response [ 16, 26 ]. Such biomarkers may be human-visible and quantifiable by manual, semi-automated, or automated means.

One such example in the field of autosomal dominant polycystic kidney disease is total kidney volume TKVa surrogate marker of disease progression which correlates with cyst volume and decline in eGFR [ 40 ]. An automated segmentation method based on deep learning has been developed to calculate TKV in a fast and reproducible manner, and demonstrated good agreement with TKV values calculated from manual segmentations [ 41 ].

Alternatively, in the field of computer vision, high-dimensional numeric data may advancementa extracted from radiologic images and analysed using machine or deep learning approaches to classify images and detect patterns which are not visible to the human eye. As part of the Biomarker Enterprise to Attack Diabetic Kidney Disease BEAt-DKD consortium, the prospective, multi-centre iBEAt cohort study is the largest DKD imaging study to date and aims to determine whether ultrasound and MRI renal imaging biomarkers provide insight into the heterogeneity in DKD pathogenesis and can prognosticate adverse outcomes amongst patients with type 2 DKD [ 26 ].

A key advantage of imaging over other biomarker approaches to personalise DKD management is the fact that nephropatht left and right kidneys as well as the renal cortex and medulla can be assessed independently, potentially providing more granularity into functional and structural heterogeneity amongst patients with DKD [ 26 ].

As diabetic retinopathy and DKD are closely intertwined as microvascular complications of diabetes mellitus, retinal imaging is also a potentially rich source of imaging biomarkers to inform DKD management [ 42 ]. Endothelial and microvessel dysfunction contribute to the development of DKD and premature reesarch disease amongst patients with diabetes mellitus [ 42 ].

Homology between the vasculature of the eye and the kidney suggests that inferences regarding the microvasculature of the kidney Diabefic be made from retinal imaging, providing a rationale to image accessible microvessels in the eye to improve DKD prognostication [ 42 ].

For example, retinal images were used to train and validate a deep learning algorithm which accurately predicted CKD status in community-based Asian cohorts [ 43 ]. The area under the receiver operating characteristic curve advancfments the deep learning researhc improved when considered alongside conventional CKD risk factors such as age, gender, ethnicity, diabetes mellitus, and hypertension [ 43 ].

By capturing nrphropathy vascular networks such as the choroidal circulation at near-histological resolution, the advent of optical advancementx tomography OCT constitutes a major advance in retinal imaging which has transformed ophthalmology care [ 44 ].

OCT can now also be deployed in preclinical models of retinopathy [ 44 ]. Deep learning has been coupled with OCT imaging to triage and diagnose the commonest sight-threatening retinal diseases in an automated fashion and with similar accuracy to that of expert physicians [ 45 ].

Thus, combining the imaging power of cross-sectional chorioretinal OCT imaging nehpropathy the analytical power of deep learning holds great promise as a means of developing prognostic imaging biomarkers related to adaptations of the renal microvasculature in people with diabetes mellitus [ 42 ].

Similar to radiologic images of the kidney, digitised whole slide images WSIs and transmission electron microscopy TEM images of kidney biopsies contain a wealth of data which may be optimally analysed using deep learning approaches [ 46, 47 ].

Deep learning approaches may be used to automate the extraction of descriptive and quantitative structural features from WSIs and TEM images with improved reproducibility [ 46, 47 ]. The concept of reproducibility is an important one as although an inter-pathologist intra-class correlation coefficient of 0.

Furthermore, semi- or wholly automated means of classifying DKD histologically would reduce personnel requirements and improve efficiency of assigning DKD diagnoses in routine clinical care. Thus, deep learning may support high-throughput and reproducible quantitative feature extraction adfancements experimental models of renal injury.

Furthermore, the trained convolutional neural network performed well on human samples, thereby providing a link between automated histopathological assessment across the preclinical and Diabdtic domains [ 49 ].

Indeed, a convolutional neural network was also used to segment PAS-stained kidney biopsy samples from 54 patients with DKD and classify them according to the Tervaert schema, achieving a high level of agreement with three independent pathologists [ 50 ]. In the assessment of resewrch structural features, deep learning has mainly been applied to digital pathology images thus far, although researchers have started to evaluate this strategy on TEM images with reasonable success [ 52 ].

Some of the biomedical technologies which support omics analyses are outlined in Figure 1. In many cases, the application of multiple technologies to characterize a particular molecular domain often provides complementary rather than redundant information.

: Diabetic nephropathy research advancements

Advances in the Research of Diabetic Nephropathy | Frontiers Research Topic

Whereas diabetes advocates a healthy and balanced diet, diet of a CKD or diabetic nephropathy patient is challenging and designed to delay progression of kidney damage and the associated secondary conditions such as hypertension, hyperlipidemia, uremia, etc. As food intake could be a burden on kidney function, a delicate balance between nutrition and sustainable physiological load is essential to maintain quality of life for the patient.

A common problem encountered in patients with renal failure and proteinuria is their lack of nutritional knowledge and continued adherence to traditional food choices that are rich in carbohydrate, proteins or minerals.

Since a majority of patients are dyslipidemic the only control they exercise is on limiting fat intake. Such a skewed diet places a tremendous burden on kidney function that causes further problems in disease management.

An ideal diet recommended for diabetic nephropathy patients with compromised kidney function includes a proper amount of fat to prevent malnutrition. More so when total calories coming from protein and carbohydrate intake needs to be restricted.

A total fat reduction as advised by earlier studies can be a very unhealthy practice. Thus, to achieve these goals nutritionists advice limiting saturated fatty acid consumption while taking vegetable oils and omega-rich fatty acid containing oils in moderation. Many clinical studies have highlighted the renoprotective effects of a low protein diet on DN, although protein restriction alone does not result in a positive outcome for patients [ 35 ].

Moreover, a protein-deficient diet 0. Interestingly, in animal type 2 DM models a very low protein diet VLPD improved tubulo-interstitial damage, inflammation and fibrosis, through restoration of autophagy via reduction of a mammalian target of rapamycin complex 1 mTORC1 activity [ 37 ].

Although a low protein diet slows progression of renal dysfunction in human subjects with chronic glomerular nephritis, VLPD has not been clinically validated. A low-salt diet that is devoid of salted and pickled foods is highly recommended for DN patients. Restricted sodium intake allows better blood pressure control in such patients.

High salt intake and urinary protein excretion were associated with annual creatinine clearance decline in type 2 DKD patients as reported by Kanauchi et al. Potassium is an essential electrolyte involved in the contraction and relaxation of muscles. During a deficit in kidney function potassium excretion is reduced leading to an accumulation in body tissues.

Therefore, potassium intake specifically from foods such as grains, potatoes, corn, soybean, nuts, tomatoes, banana, melons, kiwi etc.

must be restricted. Like potassium, phosphorus excretion is also reduced during chronic kidney damage leading to increased blood phosphorus levels. Since phosphate is in homeostatic equilibrium with the skeletal muscle calcium levels, an imbalance leads to a significant calcium loss and debilitating bone disease.

In summary, excessive carbohydrate and protein intake is managed with a target of kcal of energy per day in which 60 percent comes from carbohydrate and 40 percent from proteins.

In a recent study, such a regimen achieved a commendable control in blood lipid and glucose values in a patient with stage 4 chronic kidney disease [ 39 ]. However, patient adherence to the recommended diet seems to be gender-specific. For example, Ahola et al. Therefore, effective adherence through patient education may be a crucial factor in the management of DN through diet.

In conclusion, this review summarizes the recent advances in the pathophysiology of diabetic nephropathy and the importance of dietary factors in modifying treatment outcomes for patients.

A critical analysis of studies that emphasize the importance of patient-centered dietary intervention in successful management of advanced CKD patients has been presented. Large-scale cohort studies are necessary to evaluate the efficiency of diet as a new therapeutic paradigm.

Notably, in newly diagnosed DN patients these dietary interventions may no longer be regarded as complementary measures but significant factors that delay progression of the disease. International Diabetes Federation IDF Diabetes Atlas.

International Diabetes Federation. Andersen AR, Christiansen JS, Andersen JK, Kreiner S, Deckert T. Diabetic nephropathy in type 1 insulin-dependent diabetes: an epidemiological study. Article CAS Google Scholar.

Zhang J, Liu J, Qin X. Advances in early biomarkers of diabetic nephropathy. Rev Assoc Med Bras. Article Google Scholar.

Magee C, Grieve DJ, Watson CJ, Brazil DP. Diabetic nephropathy: a tangled web to unweave. Cardiovasc Drugs Ther. Papadopoulou-Marketou N, Paschou SA, Marketos N, Adamidi S, Adamidis S, Kanaka-Gantenbein C. Diabetic nephropathy in type 1 diabetes.

Minerva Med. PubMed Google Scholar. Nelson RG, Bennett PH, Beck GJ, et al. Diabetic Renal Disease Study Group: development and progression of renal disease in Pima Indians with non-insulin dependent diabetes mellitus.

N Engl J Med. American Diabetes Association. Nephropathy in diabetes Position Statement. Diabetes Care. Google Scholar. Ballantyne FC, Gibbons J, O-Reilly DS. Urine albumin should replace total protein for the assessment of glomerular proteinuria. Ann Clin Biochem. Kim SS, Song SH, Kim IJ, Jeon YK, Kim BH, et al.

Nonalbuminuric proteinuria as a biomarker for tubular damage in early development of nephropathy with type 2 diabetic patients. Diabetes Metab Res Rev.

Vitova L, Tuma Z, Moravec J, Kvapil M, Matejovic M, Mares J. Early urinary biomarkers of diabetic nephropathy in type 1 diabetes mellitus show involvement of kallikrein-kinin system.

BMC Nephrol. Tervaert TW, Mooyaart AL, Amann K, Cohen AH, Cook HT, Drachenberg CB, et al. Pathologic classification of diabetic nephropathy. J Am Soc Nephrol. Mogensen CE. The natural history of type 2 diabetic nephropathy. Am J Kidney Dis.

Gheith O, Farouk N, Nampoory N, Halim MA, Al-Otaibi T. Diabetic kidney disease: world wide difference of prevalence and risk factors. J Nephropharmacol. Klemens R, Angela G, Sabine H, et al. Diabetic nephropathy in 27, children, adolescents, and adults with type 1 diabetes: effect of diabetes duration, A1C, hypertension, dyslipidemia, diabetes onset and sex.

Eberhard R. Diabetic nephropathy. Saudi J Kidney Dis Transplant. Hameed I, Masoodi SR, Malik PA, Mir SA, Ghazanfar K, Ganai BA. Genetic variations in key inflammatory cytokines exacerbates the risk of diabetic nephropathy by influencing the gene expression.

Kato M, Natarajan R. Diabetic nephropathy—emerging epigenetic mechanisms. Nat Rev Nephrol. Zheng Z, Zheng F. Immune cells and inflammation in diabetic nephropathy. J Diabetes Res. Article CAS PubMed Google Scholar. Ni WJ, Tang LQ, Wei W. Research progress in signaling pathway in diabetic nephropathy.

Kawanami D, Matoba K, Utsunomiya K. Signaling pathways in diabetic nephropathy. Histol Histopathol. CAS PubMed Google Scholar. Caramori ML, Fioretto P, Mauer M. The need for early predictors of diabetic nephropathy risk: is albumin excretion rate sufficient?

Parving HH, Oenboll B, Syendsen PA, Christiansen JS, Andersen AR. Early detection of patients at risk of developing diabetic nephropathy: a longitudinal study of urinary albumin excretion.

Acta Endocrinol Copenh. Viberti GC, Hill RD, Jarrett RJ, Argyropoulos A, Mahmud U, Keen H. Microalbuminuria as a predictor of clinical nephropathy in insulin-dependent diabetes mellitus. de Boer IH, Afkarian M, Rue TC, Cleary PA, Lachin JM, Molitch ME, et al. Renal outcomes in patients with type 1 diabetes and macroalbuminuria.

Hovind P, Tarnow L, Rossing P, Jensen BR, Graae M, Torp I, et al. Predictors for the developmental of microalbuminuria and macroalbuminuria in patients with type 1 diabetes: inception cohort study. Retnakaran R, Cull CA, Thorne KI, Adler AI, Holman RR.

Risk factors for renal dysfunction in type 2 diabetes: UK prospective diabetes study Pavkov ME, Knowler WC, Bennett PH, Looker HC, Krakoff J, Nelson RG. Increasing incidence of proteinuria and declining incidence of end-stage renal disease in diabetic Pima Indians. Kidney Int. MacIssac RJ, Ekinci EI, Jerums G.

Markers of and risk factors for the development of diabetic kidney disease. Motawi TK, Shehata NI, ElNokeety MM, El-Emady YF. Potential serum biomarkers for early detection of diabetic nephropathy. Diabetes Res Clin Pract. Papadopoulou-Marketou N, Kanaka-Gantenbein C, Marketos N, Chrousos GP, Papassotiriou I.

Biomarkers of diabetic nephropathy: a update. Crit Rev Clin Lab Sci. Oltean S, Coward R, Collino M, Baelde H. Diabetic nephropathy: novel molecular mechanisms and therapeutic avenues. Biomed Res Intl. Montero RM, Covic A, Gnudi L, Goldsmith D.

Diabetic nephropathy: what does the future hold? Int Urol Nephrol. Lytvyn Y, Bjornstad P, Pun N, Cherney DZ. New and old agents in the management of diabetic nephropathy.

Curr Opin Nephrol Hypertens. Meloni C, Tatangelo P, Cipriani S, Rossi V, Suraci C, Tozzo C, et al. Adequate protein dietary restriction in diabetic and nondiabetic patients with chronic renal failure. J Ren Nutr. Otoda T, Kanasaki K, Koya D. Low-protein diet for diabetic nephropathy.

Curr Diab Rep. Trimeche A, Selmi Y, Ben Slama F, Ben Amara H, Hazar I, Ben Mami F, et al. Effect of protein restriction on renal function and nutritional status of type 1 diabetes at the stage of renal impairment.

Tunis Med. Kitada M, Ogura Y, Monno I, Koya D. A low-protein diet for diabetic kidney disease: its effect and molecular mechanism, an approach from animal studies. Kanauchi N, Ookawara S, Ito K, Mogi S, Yoshida I, Kakei M, et al.

Factors affecting the progression of renal dysfunction and the importance of salt restriction in patients with type 2 diabetic kidney disease. Clin Exp Nephrol. Kim HY. Nutritional intervention for a patient with diabetic nephropathy.

Clin Nutr Res. Ahola KAJ, Forsblom C, Groop PH. This explains, in part, the renoprotective role of SGLT2 inhibitors under conditions of normal kidney function. By blocking glucose reabsorption at the proximal tubule and diverting it into the urine, tubuloglomerular balance is restored, with the net effect of lowering intraglomerular pressure and reducing hyperfiltration 44 , Hence, the mechanism of renoprotection in advanced kidney disease is unclear.

Note that these mechanisms have not been studied in type 1 diabetes. Hyperglycemia, insulin resistance, and dyslipidemia commonly coexist, which sets in motion several dysregulated metabolic pathways inextricably related to oxidative stress and inflammatory processes, ultimately creating a vicious cycle where one process potentiates another 47 , Most notable are the polyol and protein kinase C PKC pathways, which augment oxidative stress and deplete endothelial nitric oxide synthase, respectively, leading to higher endothelin-1 and vascular endothelial growth factor levels.

Endothelial instability and nuclear factor-κB NF-κB —mediated cytokines tumor necrosis factor-α [TNF-α] and interleukin-6 [IL-6] favor an inflammatory response.

The hyperglycemic milieu also encourages the accumulation of advanced glycation end products AGEs. AGEs are a heterogeneous group of nonenzymatically glycated molecules. Upon engaging with their receptors RAGEs , which are found throughout the kidney, they trigger cellular function perturbations, including NF-κB upregulation, which induces a cascade of proinflammatory cytokines TNF-α and IL AGEs reduce the bioavailability of endothelium-derived nitric oxide and increase reactive oxygen species production, which is linked to impaired vasodilatation in diabetes 49 Fig.

Inflammation and fibrosis are major interrelated contributors to DKD progression. Mounting evidence implicates an intricate interaction between the mineralocorticoid receptor MR aldosterone and RAS-related C3 botulinum toxin substrate 1 Rac1 in driving the inflammatory processes that lead to the final common pathway of fibrosis in DKD.

The deleterious effects of MR activation in modulating inflammation and fibrosis were long recognized in animal studies of the heart and provide the therapeutic basis for MR antagonism 50 , Animal studies showed similar benefits of MR blockade on the kidneys 52 and vasculature MR activation occurs in the aldosterone-responsive distal nephron, causing sodium reabsorption and potassium excretion.

Aldosterone secretion is stimulated by RAS activation in response to decreased circulating plasma volume or significant increases in serum potassium levels. While critical for survival in states of low sodium intake, it becomes pathologic in the setting of persistently high sodium intake 54 , as exemplified by Western and many Asian diets.

Inappropriate aldosterone signaling combined with high sodium intake results in hypertension, a direct contributor to glomerular injury and fibrosis. The MR possesses a binding affinity for cortisol and corticosterone similar to that of aldosterone.

These cells typically coexpress 11B-dehydrogenase isoenzyme 2 11B-HSD2 , which neutralizes cortisol and thereby mitigates MR overactivation. Outside the distal nephron, MRs are expressed on other cell types, including podocytes, fibroblasts, vascular cells, and macrophages; these cells, however, do not uniformly coexpress the steroid-blunting effects of 11B-HSD2, which permits unbridled MR activation MR is upregulated in hyperglycemia, insulin resistance, dyslipidemia, and obesity.

This results in increased gene transcription of profibrotic factors plasminogen activator inhibitor-1 PAI-1 and TGF-β1, connective tissue growth factor, and extracellular matrix proteins, all of which contribute to progressive DKD 56 Fig.

Innate immunity plays a critical role in the pathogenesis of DKD, but a detailed discussion is beyond the scope of this review. Briefly, macrophage infiltration has been identified as one of the hallmarks of DKD, the burden of which is associated with worse disease Hyperglycemia, endothelial cell dysfunction, angiotensin II, AGEs, and oxidized LDL recruit macrophages Macrophage-MR activation polarizes macrophage differentiation toward the M1 phenotype, which promotes inflammation via a cascade of injurious cytokines 59 Figs.

Of note, however, is that MR inhibition favors macrophage switching toward an M2 anti-inflammatory phenotype with demonstrated beneficial effects in CKD The therapeutic effects of MR antagonism in curtailing DKD were first reported in using animal models There are no outcome trials with the steroidal MRAs due to tolerability issues.

However, they have been shown in people with early DKD to reduce albuminuria and blood pressure significantly. Other NS-MRAs, namely, finerenone, esaxerenone, and apararenone, also have demonstrated significant albuminuria reduction and a very low adverse effect profile in advanced DKD 64 , 65 , although esaxerenone is only available in Japan None of these agents have ever been tested in outcome trials.

The timeline of the major therapeutic outcome trials focused on delaying DKD in patients with type 2 diabetes and CKD following the publication of RENAAL and IDNT 73 — MoAs, mechanisms of action. DKD management has evolved over the last 50 years. However, following the trials that showed a clear benefit of ARBs in slowing DKD progression, nothing was proven to further curtail advancing DKD until the advent of SGLT2 inhibitors.

Many trials using novel targeted therapies were attempted between and , all of which failed to show any benefit in changing the trajectory of DKD Fig.

Since the approval of the first SGLT2 inhibitor, dapagliflozin, in January , the field has added other drugs to this class. Additionally, approval of the NS-MRA finerenone, in July , has further advanced the field from one to three therapies in the tool kit. Questions remain concerning when and how to use these drugs.

Obesity reduces the production of adiponectin in favor of leptin. Gene transcription of inflammatory mediators such as IL-1, IL-6, IL-7, IL-8, and TNF-α is increased, which creates a proinflammatory state and oxidative stress.

Profibrotic factors plasminogen activator inhibitor-1 PAI-1 and TGF-β1, connective tissue growth factor, and extracellular matrix proteins are increased, all contributing to progressive diabetic nephropathy. IL, interleukin; TNF-α, tumor necrosis factor-α; MCP-1, monocyte chemoattractant protein Many trials have established that a reduction in albuminuria is associated with slowed DKD progression and reduced CV event rates 69 — Given the advent of many new therapies over the past decade, we put forth the concept of pillars of therapy, originally adopted by heart failure cardiologists.

The concept is akin to any building structure where no single beam alone can support its standing. Hence, we now have three established, proven therapies that, when used together, will maximally decelerate DKD progression. This does not imply that those with high albuminuria, formerly called microalbuminuria, do not derive benefit from treatment—in fact, they do, and the benefits are predominantly CV.

However, the slowing of the DKD trajectory in these cohorts was detected in post hoc analyses. This ushered in two additional landmark trials conducted in patients with type 1 diabetes, Reduction of Endpoints in NIDDM with the Angiotensin II Antagonist Losartan RENAAL 20 and Irbesartan Diabetic Nephropathy Trial IDNT 19 , before RAS blockers became formally integrated into the standard of care The introduction of SGLT2 inhibitors in January created a resounding paradigm shift and renewed excitement in improving DKD management.

Although they were originally designed to lower glucose by promoting urinary glucose excretion, it was ultimately appreciated that SGLT2 inhibitors were, in effect, cardiorenal risk—reducing agents The renoprotective benefits of SGLT2 inhibitors were initially gleaned from secondary data analyses of trials with time to major adverse CV events as primary outcomes.

In a meta-analysis of six double-blinded randomized trials of SGLT2 inhibitors in patients with type 2 diabetes, a consistent reduction in hospitalization for heart failure and progression to ESKD was found However, in three landmark trials that followed, where kidney outcomes were the defined primary end points in a high-risk cohort, the benefits of SGLT2 inhibitors when combined with optimized RAS blockade became universally accepted.

The Canagliflozin and Renal Events in Diabetes with Established Nephropathy Clinical Evaluation CREDENCE trial was the first nephropathy outcome trial to specify its primary outcome as ESKD, doubling of creatinine level, or death from renal or CV causes with an SGLT2 inhibitor.

The unequivocal renal benefits prompted early trial termination The Dapagliflozin and Prevention of Adverse Outcomes in Chronic Kidney Disease DAPA-CKD trial, which was published a year later than the CREDENCE trial, randomized a mixed cohort of participants, i.

Most recently, The Study of Heart and Kidney Protection With Empagliflozin EMPA-KIDNEY outcome trial adds to the substantial body of evidence underscoring the robust salutary effects of SGLT2 inhibitors on renal outcomes.

Over 6, participants with and without diabetes, with a wide range of GFR declines, from mild to severe, and with normal or high albuminuria, were randomized to empagliflozin versus placebo in addition to standard-of-care therapies.

It should further be noted that there were many people with normal and high albuminuria in this trial. Hence, SGLT2 inhibitors serve as a solid second pillar to slow DKD progression The hazardous sequela of MR activation by aldosterone shifted attention to developing therapies downstream of the RAS target, which ultimately addresses the inflammatory and ensuing profibrotic processes that contribute to DKD progression and heart failure.

MR antagonism is not new. Steroid-based MRAs, including first- and second-generation spironolactone and eplerenone, respectively, continue to be used extensively in symptomatic heart failure patients This has been adopted by the U.

Food and Drug Administration as evidence of benefit and as a guideline by the American Diabetes Association. The exception is when a dual RAS blockade is used. Therefore, MRAs have been generally contraindicated in advanced kidney disease Finerenone, apararenone, esaxerenone, and ocedurenone are members of a new class of NS-MRAs with pharmacologic properties distinct from those of their distant cousins, the steroidal agents.

Finerenone is the only one developed and approved for cardiorenal risk reduction, whereas the others are approved only for blood pressure control, with no outcome data supporting use in DKD.

Finerenone demonstrates superior anti-inflammatory and antifibrotic outcomes compared with its steroidal counterparts in preclinical studies 92 — Unlike the steroidal MRAs, the NS-MRAs achieve balanced tissue distribution between the heart and kidney rather than affecting the kidney alone Finerenone was studied in two complementary phase 3 randomized, double-blinded, placebo-controlled clinical trials that included over 13, participants with type 2 diabetes optimized on maximally tolerated RAS blockade before randomization to the NS-MRA or placebo 96 , These trials were developed with the same protocol but different inclusion criteria, which allowed for an individual pooled patient analysis in the Finerenone in Chronic Kidney Disease and Type 2 Diabetes: Combined FIDELIO-DKD and FIGARO-DKD Trial Programme Analysis FIDELITY Despite the similar frequency of adverse effects in both groups, the incidence of trial discontinuation related to hyperkalemia was slightly higher with finerenone than with placebo 2.

The frequency of reported adverse effects was similar to that for placebo, and the incidence of trial discontinuation related to hyperkalemia was slightly higher with finerenone than with placebo 1.

The FIDELITY individual pooled patient analysis included 13, patients with type 2 diabetes and a wide range of CKD stages, from 1 to 4, and high to very high albuminuria. This is a very important finding given the notably higher risk of heart failure and premature death associated with having both diabetes and CKD 4 , 99 , Studies of finerenone in type 1 diabetes are currently underway.

The incidence of clinically important adverse effects related to hyperkalemia was slightly higher for finerenone, with 1. However, unlike many other trials, all participants were required to be on maximally tolerated RAS blockade.

Differences in hyperkalemia risk between the NS-MRA finerenone and its steroidal counterpart, spironolactone, are exemplified by a comparative outcome of a subgroup with resistant hypertension from the FIDELITY analysis and the Spironolactone With Patiromer in the Treatment of Resistant Hypertension in Chronic Kidney Disease AMBER trial In the FIDELITY study, the incidence of hyperkalemia was GLP-1 RAs are recommended for patients with DKD who have not met their glycemic targets despite optimization with metformin and SGLT2 inhibitors Post hoc analyses of these CVOT, like analyses of SGLT2 inhibitors, also demonstrated possible benefits in delaying DKD progression Additionally, a large analysis of more than 12, participants pooled from the earliest trials of GLP-1 RAs, Trial to Evaluate Cardiovascular and Other Long-term Outcomes With Semaglutide in Subjects With Type 2 Diabetes SUSTAIN-6 and Liraglutide Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results LEADER , evaluated changes in albuminuria, rate of annual eGFR change, and time to persistent eGFR declines.

Semaglutide and liraglutide were associated with significant slowing of annual eGFR decline, 0. This trial enrolled 4, patients with known type 2 diabetes and either current CKD eGFR Notably, there was a higher prevalence of CKD in the trial cohort than in the seven CVOT studies completed to date.

Additionally, Since Abdominal obesity is independently associated with albuminuria despite normoglycemia and normotension and addresses an important subgroup of patients who have obesity-related glomerulopathy and who, by conventional screening, may be classified as metabolically healthy yet are still at risk for developing ESKD This is not surprising given the inflammatory milieu engendered by excess adiposity Fig.

FLOW A Research Study to See How Semaglutide Works Compared to Placebo in People With Type 2 Diabetes and Chronic Kidney Disease is an ongoing randomized placebo-controlled trial that assesses the efficacy of semaglutide in type 2 diabetes and CKD and the first designed with primary renal end points The renoprotective properties associated with GLP-1 RAs reviewed here were drawn from post hoc analyses of major CVOT, the evidence of which has been compelling, nonetheless.

DKD management should start with maximally dosed RAS blockade; this is based on the doses used in the original RAS blocking trials, wherein dose reduction to avoid hyperkalemia resulted in markedly reduced protection against DKD decline. Each drug class that provided improved outcomes was coupled with RAS blockade and independently showed benefits on kidney and heart outcomes.

These findings provide impetus to adopt a pillared approach for reducing cardiorenal events similar to how heart failure cardiologists have approached heart failure management.

Notably, there has never been a trial that evaluated the simultaneous use of all four agents in heart failure or that compared different drug combinations against each other.

Each trial was assessed on its merits within each drug class and then combined in retrospect to provide the best results. We currently have three agents with additive effects on albuminuria and heart failure outcomes when combined individually with RAS blockade.

An adequately statistically powered study that evaluates the three, or possibly soon-to-be four, different agents together would require well over , participants; therefore, extrapolation, which the cardiologists have done successfully, uses all three agents together over a short time.

Practice guidelines articulate that clinicians should start first by titrating to maximally tolerated RAS blockade before introducing these medications SGLT2 inhibitors, NS-MRAs, and GLP-1 RAs , as was done in pivotal clinical trials Initial changes in eGFR and associated long-term amelioration of renal function decline have also been reported with SGLT2 inhibitors and finerenone, albeit to a lesser degree than with RAS blockers 96 , , — The manifold pathophysiological mechanisms involved in end-organ damage argue for a pillared approach with targeted therapies that have distinct pharmacodynamic actions An elegant study that used an animal model of preclinical hypertension—induced cardiorenal disease with a low-dose combination therapy of finerenone and empagliflozin revealed additive cardiorenal benefit above that of the respective dose-dependent monotherapy, as measured by reductions in blood pressure, proteinuria, cardiac fibrosis, vasculopathy, and mortality These findings further argue strongly for distinct pharmacodynamic actions that counteract the manifold pathophysiological mechanisms involved in end-organ damage Fig.

Acute changes in GFR slopes with three distinct classes of drugs with unique mechanisms that slow kidney disease progression associated diabetes.

B : Rate of eGFR decline between finerenone and placebo. Despite equivalent GFR at baseline, the finerenone group shows stabilization of slowed GFR declines 96 and higher GFR declines at 4 months, which are associated with better long-term outcomes.

C : Initial decline in GFR decline in the empagliflozin group compared with placebo at month 4 EMPA-REG Outcome, BI Empagliflozin Cardiovascular Outcome Event Trial in Type 2 Diabetes Mellitus Patients; LS, least squares.

Additionally, in a small open-label randomized crossover clinical study, the efficacy and safety of dapagliflozin and a low dose of the steroidal MRA eplerenone were evaluated in a cohort of patients with CKD.

The combination of the two drugs was associated with an additive effect on albuminuria reduction compared with the use of either drug alone. Importantly, the incidence of hyperkalemia was significantly less in the combination group than in the group that received eplerenone alone This is consistent with data from the larger FIDELIO-DKD trial that demonstrated greater protection from hyperkalemia when an SGLT2 inhibitor was combined with finerenone The use of combination therapies with NS-MRAs and SGLT2 inhibitors was further explored in the FIDELITY subgroup analysis, which revealed that, compared with placebo, cardiorenal benefits of finerenone were appreciably higher irrespective of concomitant GLP-1RA or SGLT2 inhibitor use at baseline or anytime during the trial.

An important caveat is that GLP-1 RA and SGLT2 inhibitor users comprised 6. More importantly, there was no sign that drug coadministration with finerenone increased any risk of kidney injury When combined with finerenone, SGLT2 inhibitors reduced hyperkalemic events compared with levels found in nonusers 8.

These are separate retrospective analyses of renal outcome trials clearly showing a protective effect of SGLT2 inhibitors from hyperkalemia in the setting of NS-MRA and MRA use 98 , Since the institution of the RAS blockade in the s, we have witnessed significant strides in addressing the unmitigated risk associated with DKD progression.

We now have two additional drug classes to add to the RAS blockers, SGLT2 inhibitors and NS-MRAs, bolstered by a robust body of outcome data, and a possible third class.

The efficacy of GLP-1 RAs is supported by retrospective analyses but needs to be proven in the ongoing FLOW randomized clinical trial. The safety and tolerability of these two drug classes, when given together against a backdrop of maximal RAS blockade, are very encouraging and reflect the complexity of the underlying pathophysiology that drives DKD progression.

Moreover, the protective role of SGLT2 inhibitors in preventing hyperkalemia with finerenone use is very heartening. This article is featured in a podcast available at diabetesjournals. was supported in part by a National Institutes of Health T32 award. Duality of Interest. reports being a consultant to and on steering committees of several outcome trials of companies, including KBP Biosciences, Janssen, Novo Nordisk, Ionis, Alnylam, ESTAR, Bayer, and AstraZeneca.

No other potential conflicts of interest relevant to this article were reported. Author Contributions. and G. researched the data and shared writing and editing responsibilities for the manuscript.

Sign In or Create an Account. Search Dropdown Menu. header search search input Search input auto suggest. filter your search All Content All Journals Diabetes Care. Advanced Search. User Tools Dropdown. Sign In. Skip Nav Destination Close navigation menu Article navigation.

Volume 46, Issue 9. Previous Article Next Article. Pathophysiologic Mechanisms and Potential Therapeutic Targets. Clinical Application.

The Additive Value of Drug Combination. Article Information. Article Navigation. Review August 25 Diabetic Nephropathy: Update on Pillars of Therapy Slowing Progression Sandra C. Naaman ; Sandra C. Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and American Heart Association Comprehensive Hypertension Center, University of Chicago Medicine, Chicago, IL.

This Site. Google Scholar. George L. Bakris Corresponding author: George L. Bakris, gbakris uchicago. Diabetes Care ;46 9 — Article history Received:. Get Permissions. toolbar search Search Dropdown Menu. toolbar search search input Search input auto suggest. Graphical Abstract View large Download slide.

View large Download slide. Figure 1. Figure 2. Figure 3. Figure 4. Figure 5. Figure 6. Figure 7. Albuminuria and renal insufficiency prevalence guides population screening: results from the NHANES III.

Search ADS. Worldwide epidemiology of diabetes-related end-stage renal disease, National Institute of Diabetes and Digestive and Kidney Diseases. Accessed 14 May NCD Risk Factor Collaboration NCD-RisC. Worldwide trends in diabetes since a pooled analysis of population-based studies with 4.

Frontiers | Editorial: Advances in the research of diabetic nephropathy, volume II

In addition, incretin-related drugs showed a renoprotective ability in many clinical trials, and these trials with renal outcome as their primary endpoint are currently ongoing.

Hypoxia-inducible factor prolyl hydroxylase inhibitors recently approved for renal anemia may be renoprotective since they improve tubulointerstitial hypoxia.

Thus, following SGLT2 inhibitor, numerous novel drugs could be utilized in treating DKD. Future studies are expected to provide new insights. Keywords : Diabetic nephropathies ; Epigenomics ; Glycation end products, advanced ; Hypoxia-inducible factor 1 ; NF-E2-related factor 2 ; Sodium-glucose transporter 2.

The concept of diabetic kidney disease and diabetic nephropathy. GFR, glomerular filtration rate. The time course of current treatments. The time course of future treatments. Citations Citations to this article as recorded by. PubReader ePub Link Cite CITE. export Copy Format NLM AMA APA MLA.

Download Citation Download a citation file in RIS format that can be imported by all major citation management software, including EndNote, ProCite, RefWorks, and Reference Manager.

Format: RIS — For EndNote, ProCite, RefWorks, and most other reference management software BibTeX — For JabRef, BibDesk, and other BibTeX-specific software Include: Citation for the content below Citation and abstract for the content below Treatment of Diabetic Kidney Disease: Current and Future.

Diabetes Metab J. pasue play. Sign up. Sign up for the DMJ newsletter— what matters in science, free to your inbox daily.

Simultaneously, tubular basement thickening, parallel to GBM thickening, is also observed. As for type 2 diabetes, the pathological changes are more complicated and often classified into 3 categories [ 53 ]: class I is the classic diabetic glomerulopathy as described above; class II has predominant vascular and interstitial changes, relatively normal glomerular structure.

This group of patients often has an early decline of GFR without significant albuminuria. Clinically, patients have an early decline of GFR without significant proteinuria.

Mechanistically, these patients may have unrecognized AKI episodes during the disease progression. It is unclear whether diabetes interacts with the primary glomerular disease to accelerate the progression of CKD. In type 1 diabetes, most patients could have a diagnosis of DKD based on clinical presentation.

However, the diagnosis of DKD in type 2 diabetes often requires additional tests including kidney biopsy. Diabetes is a common disease with high coincidence with other nondiabetic CKD. Another interesting observation is that clinical manifestation is not necessarily associated with histological findings in DKD, as suggested by an autopsy study [ 41 ].

In that research, of patients had histopathologic changes of DN in the kidney, but 20 of histologically proven DN cases did not present with DN-associated clinical manifestations within their lifetime. At the most clinical practice, patients with atypical clinical features should prompt evaluation for non-DKD by performing kidney biopsy.

These atypical presentations are constructed of rapidly decreasing renal function, rapidly increasing proteinuria, active urinary sediment, refractory hypertension, and nephrotic range of proteinuria with normal GFR.

Membranous diseases or minimal change disease should be ruled out from a nephrotic range of proteinuria with normal GFR because these patients could be treated with steroids and immunosuppressive therapy. Conventional therapy of DKD includes better hyperglycemic control, RAS blockers, and other managements such as lipid-lowering therapy and so on.

In humans, RAS inhibition has proved to be the single most effective therapy for slowing the progression of DN [ 54 ]. However, 3 randomized, placebo-controlled trials of —3, patients with type 1 diabetes and normoalbuminuria RASS [ 55 ], EUCLID [ 56 ], AND DIRECT [ 57 ] suggest that early therapy in patients, type 1 diabetes is ineffective in preventing the development of MA.

In addition, the combination therapy with an angiotensin-converting enzyme inhibitor plus an angiotensin II receptor blocker does not prevent renal disease progression or death, and it increases the rate of serious adverse events such as AKI, hyperkalemia, and hypotension, as shown by the Veterans Affairs Nephropathy in Diabetes study VA NEPHRON-D [ 58 ].

The continued increase in diabetic ESRD urgently requires additional innovative therapies. A better understanding of the pathogenesis of DKD could help us to identify new drug targets for DKD.

The recent success of SGLT2 inhibitors as a new therapy for DKD patients is exciting and encouraging news for the nephrologists. SGLT2 inhibitors have been shown to lower body weight, blood pressure, serum uric acid, and glomerular hyperfiltration through increased urinary excretion of glucose and sodium, osmotic diuresis, and improved tubule-glomerular feedback mechanism [ 60 ].

In patients with type 2 DM and high cardiovascular risks, empagliflozin is associated with slower progression of kidney disease and lower rates of clinically relevant renal events than placebo when added to standard care [ 62, 63 ].

Canagliflozin, another SGLT2 inhibitor, was shown in patients with type 2 diabetes and kidney disease CREDENCE ; the risk of kidney failure was significantly lower in the canagliflozin group than in the placebo group at a median follow-up of 2.

In Dekkers research, they demonstrated that 6 weeks of dapagliflozin decreased albuminuria by In another study NCT , in patients with type 2 diabetes and CKD 3A, decreases from baseline in eGFR were greater with dapagliflozin than placebo at Week 24 —2.

However, attentions should be paid to side effects such as hypoglycemia, urinary and genital infections, elevated liver enzymes, bone fractures, or amputations. Glucagon-like peptide-1 GLP-1 is one of the incretins released from the intestine in response to food intake and it can stimulate insulin secretion.

Its level is decreased and its analogs e. liraglutide have been used in type 2 DM. According to the Liraglutide Effect and Action in Diabetes: Evaluation of Cardiovascular Outcomes Results double-blind trial with 9, patients with type 2 DM and high cardiovascular risk, patients with liraglutide had lower rate of new onset of persistent macroalbuminuria and progression of DKD than placebo [ 68, 69 ].

In another randomized study of 3, patients with type 2 diabetes, patients using semaglutide another GLP-1 analog had lower rates of new or deteriorating nephropathy than those with placebo [ 70 ]. Dipeptidyl peptidase 4 inhibitors linagliptin and saxagliptin can reduce the amount of albuminuria, but the evidence is less clear than GLP-1 analog [ 71, 72 ].

Peroxisome proliferator activated receptor-γ agonist, rosiglitazone, can significantly decrease urinary albumin to creatinine ratio in type 2 diabetes patients after 3 months of therapy [ 73 ].

Avosentan, an endothelin-1 receptor A antagonist, can reduce urinary albumin excretion, but the study was terminated early because of excessive cardiovascular events during the treatment course due to the fluid overload [ 75 ].

A recent study in patients showed that atrasentan, a more selective endothelin receptor A antagonist, had a proteinuria-lowering effect without significant accumulation of body fluid [ 76 ].

However, this study was terminated early due to the recruitment issue. Future studies are required to confirm whether atrasentan has additional renal protective effects on top of the RAS blockades and SGLT2 inhibitors, the new standard care for DKD patients.

Mineralocorticoid receptor antagonists MRA exert an antifibrotic and anti-inflammatory effect on the kidneys and other target organs like the heart and vessels [ 77 ].

Due to significant side effects associated with the first- spironolactone and second- eplerenone generation MRA, new MRA such as apararenone also called MT , esaxerenone, and finerenone have a relatively low risk of hyperkalemia. A clinical trial of MT in patients with DKD NCT is ongoing in Japan clinicaltrials.

Esaxerenone is also being studied in phase II and III randomized clinical trial RCT , but no results have been published yet [ 78 ]. Finerenone has been investigated in several phase II RCTs.

In the MRA tolerability study-heart failure ARTS-HF , finerenone reduced albuminuria in patients with CKD and heart failure and had a lower risk of hyperkalemia than spironolactone [ 79 ].

The risk of hyperkalemia leading to discontinuation was 2. Two double-blind placebo-controlled phase III RCTs, FIDELIO NCT and FIGARO NCT are currently ongoing to examine the effects of finerenone on DKD and cardiovascular outcomes [ 81, 82 ].

Another phase III RCT NCT to determine the effect of pirfenidone on GFR and albuminuria in patients with DKD is still ongoing. Pentoxifylline, another nonspecific antifibrotic agent, was shown to reduce albuminuria and slow progression of renal disease in patients with type 2 diabetes with stages 3—4 CKD on top of RAS blockade [ 83 ].

Two more clinical trials NCT and NCT are ongoing. Hyperglycemia-associated generation of advanced glycation end products AGE and engagement of the receptor for AGE with its ligands can induce oxidative stress and renal inflammation. Pyridoxamine, a member of family of vitamin B6, can remove free radicals and carbonyl products and block synthesis of AGEs.

Clinical studies showed that pyridoxamine did not provide a significant renal protection in DKD patients [ 84 ], but a significant beneficial effect was observed in a subgroup of DKD population.

Therefore, more clinical studies are required to further confirm this finding. Not only in immune cells, but JAK-signal transducer and activator of transcription also play critical roles in renal cells, including mesangial cells, podocytes, and tubular epithelial cells [ 85 ].

This pathway is activated by reactive oxygen species induced by the hyperglycemic state [ 86 ]. A phase II clinical trial using Baricitinib, a selective JAK-1 and JAK-2 inhibitor, showed a reduction of proteinuria and expression of several inflammatory markers in patients with DKD [ 85 ].

Nuclear factor-2 erythroid related factor Nrf2 -keep 1 pathway has been also shown to play a major role in the progression of DKD [ 87 ].

Pharmacological activation of Nrf2 decreases cytokine production, M1 macrophage accumulation, and the formation of an atherosclerotic plaque lipid core in the experimental model of streptozotocin-induced diabetic mice on an apolipoprotein E-deficient background [ 88 ].

In addition, its activation improves the pathological changes in the glomerulus of streptozotocin-induced diabetic mice through a reduction in oxidative stress, TGF-β expression, and extracellular matrix proteins [ 89 ].

Bardoxolone methyl reduces the generation of oxidative stress by the activation of Nrf2 and inhibition of NF-kB pathway. The short-term study revealed that the use of bardoxolone methyl can increase the GFR in patients with type 2 DM and impaired renal function but have no influence of albuminuria [ 23 ].

However, the large phase III clinical study was terminated early because of more cardiovascular events [ 90 ]. Oxidative stress plays a critical role in the pathogenesis of DN [ 91 ].

NADPH oxidase NOX enzyme isoforms are involved in the production of reactive oxygen species that cause kidney cell injury in DKD [ 92 ]. However, the phase II trial in patients with T2DM and albuminuria failed to show any significant renal protection [ 93 ].

APX, a pan-NOX inhibitor, has been shown to have a renal protective effect in an experimental animal model of diabetes [ 94 ], but human studies have not been done yet. The proinflammatory chemokine ligand 2, also known as MCP-1, has been implicated in the pathogenesis of DN and has become a novel treatment target.

Mouse models treated with NOX-E36, a chemokine ligand 2 inhibitor, exhibited a reduction in albuminuria [ 95 ]. A phase II clinical trial demonstrated that treatment of NOX-E36 reduced albuminuria in patients with T2DM and DN [ 96 ].

Long-term effects of these medications on renal outcomes and mortality are still in need. All drugs for DKD treatment are shown in Table 1. DKD remains the most common cause of ESRD in the world including the US and China.

DM patients with CKD could have a variety of clinical presentations with or without albuminuria. We need to perform more kidney biopsy for DKD patients with albuminuria because a significant number of DM patients could have non-DKD glomerular disease.

The clinical presentation does not necessarily correlate with histological changes in DKD patients. Identification of biomarkers for early detection of DKD patients at high risk for progression could help us to better manage DKD patients and prevent their progression. Identification of SGLT2 inhibitors as a new therapy for DKD has a huge impact, however, is not sufficient to halt the progression of DKD.

Therefore, there is an urgent need to better understand the pathogenesis of DKD and develop more drugs to treat these patients. is supported by VA Merit Award, NIH 1R01DK, NIH 1R01DK, NIH PDK; K. is supported by NIH R01 DK Sign In or Create an Account. Search Dropdown Menu.

header search search input Search input auto suggest. filter your search All Content All Journals Kidney Diseases. Advanced Search. Skip Nav Destination Close navigation menu Article navigation. Volume 6, Issue 4. Good and Bad News in the DKD Epidemiology. Albuminuria Is Not Necessarily Associated with a Decline in Glomerular Filtration Rate in DKD.

Identification of High-Risk DKD Patients. New Insights into DKD Pathology. Challenges and Opportunities in Developing New Therapies for DKD. Disclosure Statement. Funding Sources.

Author Contributions. Article Navigation. Review Articles March 31 Diabetic Kidney Disease: Challenges, Advances, and Opportunities Subject Area: Nephrology. Ya Chen ; Ya Chen. b Department of Nephrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.

This Site. Google Scholar. Kyung Lee ; Kyung Lee. Zhaohui Ni ; Zhaohui Ni. John Cijiang He John Cijiang He. he mssm.

Kidney Dis 6 4 : — Article history Received:. Cite Icon Cite. toolbar search Search Dropdown Menu. toolbar search search input Search input auto suggest. View large Download slide. Table 1. Review of drugs for DKD treatments.

View large. View Large. There are no acknowledgments to declare. The authors have no conflicts of interest to declare. and J. wrote the manuscript; K. and Z. revised manuscript. Karger Publishers [Internet].

Centers for Disease Control and Prevention: National Diabetes Statistics Report, Prevalence and Ethnic Pattern of Diabetes and Prediabetes in China in Search ADS. Global aetiology and epidemiology of type 2 diabetes mellitus and its complications.

How to prevent the microvascular complications of type 2 diabetes beyond glucose control. Incidence of End-Stage Renal Disease Attributed to Diabetes Among Persons with Diagnosed Diabetes - United States and Puerto Rico, In patients with type 1 diabetes and new-onset microalbuminuria the development of advanced chronic kidney disease may not require progression to proteinuria.

Progressive renal decline: the new paradigm of diabetic nephropathy in type 1 diabetes. The Presence and Consequence of Nonalbuminuric Chronic Kidney Disease in Patients With Type 1 Diabetes. Microalbuminuria and the risk for early progressive renal function decline in type 1 diabetes.

Normoalbuminuric renal-insufficient diabetic patients: a lower-risk group. Diverging association of reduced glomerular filtration rate and albuminuria with coronary and noncoronary events in patients with type 2 diabetes: the renal insufficiency and cardiovascular events RIACE Italian multicenter study.

Clinical Manifestations of Kidney Disease Among US Adults With Diabetes, Renal insufficiency in the absence of albuminuria and retinopathy among adults with type 2 diabetes mellitus.

Non-proteinuric pathways in loss of renal function in patients with type 2 diabetes. Rate and determinants of association between advanced retinopathy and chronic kidney disease in patients with type 2 diabetes: the Renal Insufficiency And Cardiovascular Events RIACE Italian multicenter study.

de Boer. Intensive diabetes therapy and glomerular filtration rate in type 1 diabetes. Acute kidney injury episodes and chronic kidney disease risk in diabetes mellitus. Acute Kidney Injury Predicts Major Adverse Outcomes in Diabetes: Synergic Impact With Low Glomerular Filtration Rate and Albuminuria.

The influence of multiple episodes of acute kidney injury on survival and progression to end stage kidney disease in patients with chronic kidney disease. Risk of end-stage renal disease in diabetes mellitus: a prospective cohort study of men screened for MRFIT.

Multiple Risk Factor Intervention Trial. The growing volume of diabetes-related dialysis: a population based study. Heterogeneity of diabetes outcomes among asians and pacific islanders in the US: the diabetes study of northern california DISTANCE.

Rates of complications and mortality in older patients with diabetes mellitus: the diabetes and aging study. Cumulative Risk of End-Stage Renal Disease Among Patients With Type 2 Diabetes: A Nationwide Inception Cohort Study.

Role of intensive glucose control in development of renal end points in type 2 diabetes mellitus: systematic review and meta-analysis intensive glucose control in type 2 diabetes.

Persistent renal hypertrophy and faster decline of glomerular filtration rate precede the development of microalbuminuria in type 1 diabetes.

The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. Association of HbA1c levels with vascular complications and death in patients with type 2 diabetes: evidence of glycaemic thresholds.

Structural Predictors of Loss of Renal Function in American Indians with Type 2 Diabetes. Insights to the genetics of diabetic nephropathy through a genome-wide association study of the GoKinD collection.

Plasma Biomarkers and Kidney Function Decline in Early and Established Diabetic Kidney Disease. Cellular basis of diabetic nephropathy: 1. Study design and renal structural-functional relationships in patients with long-standing type 1 diabetes.

The early natural history of nephropathy in type 1 diabetes: II. Early renal structural changes in type 1 diabetes. The early natural history of nephropathy in type 1 diabetes: I.

Topic Editors Of note, however, nepgropathy that MR inhibition advancenents macrophage switching Diabetic nephropathy research advancements an M2 anti-inflammatory Lowering AC levels with demonstrated beneficial effects in CKD As animal gesearch do Diabetic nephropathy research advancements reliably recapitulate all features of human Dizbetic [ 23 ], access to human kidney tissue from patients with DKD and from healthy controls is essential. de Boer IH, Alpers CE, Azeloglu EU, Balis UGJ, Barasch JM, Barisoni L, et al. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. Initial eGFR changes with ertugliflozin and associations with clinical parameters: analyses from the VERTIS CV Trial.
Top bar navigation

Some of the biomedical technologies which support omics analyses are outlined in Figure 1. In many cases, the application of multiple technologies to characterize a particular molecular domain often provides complementary rather than redundant information.

Moreover, integration of data from several molecular domains is key to characterizing the molecular heterogeneity of DKD, although integrative multi-omic analyses are not trivial owing to the complexity of the multiple high-dimensional datasets involved [ 17, 56 ]. It is also worth noting that changes in different molecular domains such as the transcriptome, the proteome, and the metabolome may not necessarily directly correlate [ 16, 57, 58 ].

For example, factors impacting translational efficiency will diminish mRNA-protein correlations for a given target, as will modalities of protein regulation other than gene transcription, such as post-translational modifications [ 57 ].

Furthermore, differences in the coverage of molecular domains by omics technologies may result in difficulties mapping insights from one to the other [ 16, 58 ].

Techniques that allow for integration of not only two data domains at a time such as the transcriptome and the proteome but also allow for simultaneous integration of clinical phenotypic data, imaging data, and histopathological data along with multiple molecular omics data domains are essential to gain more holistic insights into cellular function and interaction in a complex organ system such as the diabetic kidney [ 17, 56 ].

The current one-size-fits-all approach to DKD care ignores the clinically apparent heterogeneity in disease prognosis and treatment-responsiveness [ 10 ]. It is hoped that a systems biology approach to DKD research will pave the way for a precision medicine approach to routine DKD care by unravelling individual-level molecular mechanisms which underlie progressive DKD and which are amenable to targeting by existing or novel therapeutic strategies [ 15, 16 ].

Certain priorities for translational DKD research which may be advanced by a systems nephrology approach include: 1. The development of model systems in vitro or animal which reliably recapitulate progressive and advanced human DKD characterized by single-cell and spatially resolved transcriptomics, thereby enhancing the translational relevance of preclinical DKD studies;.

The identification of biomarkers which predict response to RAAS blockade, SGLT2is, and other emerging disease-modifying treatments for DKD in light of the inter-individual variability in treatment response; and. The delineation of mechanisms of DKD progression in the face of combined therapy with RAAS blockade and an SGLT2i, the current backbone of treatment, which may help define targets for novel therapies which minimise the significant residual risk of progressive renal functional decline.

However, the efficacy of appropriately targeted novel therapeutics may still be impacted by inter-individual pharmacokinetic differences.

Thus, pharmacogenomic profiling will play an important role in optimising outcomes for individuals with DKD. While a comprehensive systems nephrology approach is now technically feasible in research studies, this must be balanced with plans for eventual implementation of elements of this paradigm in clinical practice.

The value of biological insights derived from the refined techniques currently available must be balanced against their clinical translatability; researchers and clinicians alike should grapple with this compromise from the outset in an effort to prioritize which elements of the systems nephrology paradigm offer benefit to the largest number of patients in clinical practice.

This will help to ensure that implementation of a systems nephrology approach in routine DKD care will not perpetuate, or indeed exacerbate, inequity in healthcare delivery. This manuscript was invited following a presentation at the European Renal Association Diabesity Working Group Annual CME in Maribor, Slovenia on September 16th—17th Figure 1 was created with BioRender.

This work was performed within the Irish Clinical Academic Training ICAT Programme, supported by the Wellcome Trust and the Health Research Board Grant No. This research was funded in whole, or in part, by the Wellcome Trust Grant No. For the purpose of open access, the author has applied a CC BY public copyright licence to any Author Accepted Manuscript version arising from this submission.

Martin wrote the manuscript and Neil G. Docherty provided proof-reading and critical review. Martin and Neil G. Docherty reviewed and approved the final manuscript. Sign In or Create an Account. Search Dropdown Menu. header search search input Search input auto suggest. filter your search All Content All Journals Nephron.

Advanced Search. Skip Nav Destination Close navigation menu Article navigation. The Promise of Personalised Diabetic Kidney Disease Care. Model Systems and Data Sources. Non-Omics Data. Molecular Omics Data. Conflict of Interest Statement. Funding Sources. Author Contributions. Article Navigation.

Review Articles September 11 Early Publication. A Systems Nephrology Approach to Diabetic Kidney Disease Research and Practice Subject Area: Nephrology. Martin Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.

liampvmartin gmail. This Site. Google Scholar. Neil G. Docherty Neil G. Nephron 1— Article history Received:. Cite Icon Cite. toolbar search Search Dropdown Menu. toolbar search search input Search input auto suggest. Journal Section:.

View large Download slide. Table 1. An essential toolkit for a systems nephrology approach to diabetic kidney disease a. View Large. The development of model systems in vitro or animal which reliably recapitulate progressive and advanced human DKD characterized by single-cell and spatially resolved transcriptomics, thereby enhancing the translational relevance of preclinical DKD studies; 2.

The identification of biomarkers which predict response to RAAS blockade, SGLT2is, and other emerging disease-modifying treatments for DKD in light of the inter-individual variability in treatment response; and 3. The authors have no conflicts of interest to declare.

Oshima M, Shimizu M, Yamanouchi M, Toyama T, Hara A, Furuichi K, et al. Trajectories of kidney function in diabetes: a clinicopathological update. Nat Rev Nephrol.

Afkarian M, Sachs MC, Kestenbaum B, Hirsch IB, Tuttle KR, Himmelfarb J, et al. Kidney disease and increased mortality risk in type 2 diabetes. J Am Soc Nephrol. Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, et al.

Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med. Heerspink HJL, Stefánsson BV, Correa-Rotter R, Chertow GM, Greene T, Hou F-F, et al. Dapagliflozin in patients with chronic kidney disease.

Shaman AM, Bain SC, Bakris GL, Buse JB, Idorn T, Mahaffey KW, et al. Effect of the glucagon-like peptide-1 receptor agonists semaglutide and liraglutide on kidney outcomes in patients with type 2 diabetes: pooled analysis of SUSTAIN 6 and LEADER.

Heerspink HJL, Sattar N, Pavo I, Haupt A, Duffin KL, Yang Z, et al. Effects of tirzepatide versus insulin glargine on kidney outcomes in type 2 diabetes in the SURPASS-4 trial: post-hoc analysis of an open-label, randomised, phase 3 trial.

Lancet Diabetes Endocrinol. Bakris GL, Agarwal R, Anker SD, Pitt B, Ruilope LM, Rossing P, et al. Effect of finerenone on chronic kidney disease outcomes in type 2 diabetes. Martin WP, Docherty NG, Le Roux CW. Impact of bariatric surgery on cardiovascular and renal complications of diabetes: a focus on clinical outcomes and putative mechanisms.

Expert Rev Endocrinol Metab. Martin WP, White J, Lopez-Hernandez FJ, Docherty NG, le Roux CW. Metabolic surgery to treat obesity in diabetic kidney disease, chronic kidney disease, and end-stage kidney disease; what are the unanswered questions? Front Endocrinol. Tye SC, Denig P, Heerspink HJL.

Precision medicine approaches for diabetic kidney disease: opportunities and challenges. Nephrol Dial Transplant. Luyckx VA, Rule AD, Tuttle KR, Delanaye P, Liapis H, Gandjour A, et al. Nephron overload as a therapeutic target to maximize kidney lifespan. Rossing P, Caramori ML, Chan JCN, Heerspink HJL, Hurst C, Khunti K, et al.

KDIGO clinical practice guideline for diabetes management in chronic kidney disease. Kidney Int. Krolewski AS. Progressive renal decline: the new paradigm of diabetic nephropathy in type 1 diabetes.

Diabetes Care. de Boer IH, Alpers CE, Azeloglu EU, Balis UGJ, Barasch JM, Barisoni L, et al. Rationale and design of the kidney precision medicine project.

Mariani LH, Pendergraft WFIII, Kretzler M. Defining glomerular disease in mechanistic terms: implementing an integrative biology approach in nephrology.

Clin J Am Soc Nephrol. Lindenmeyer MT, Alakwaa F, Rose M, Kretzler M. Perspectives in systems nephrology. Cell Tissue Res. Eddy S, Mariani LH, Kretzler M. Integrated multi-omics approaches to improve classification of chronic kidney disease.

Pushpakom S, Iorio F, Eyers PA, Escott KJ, Hopper S, Wells A, et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov. Tuttle KR, Brosius FCIII, Adler SG, Kretzler M, Mehta RL, Tumlin JA, et al. Hence, the mechanism of renoprotection in advanced kidney disease is unclear.

Note that these mechanisms have not been studied in type 1 diabetes. Hyperglycemia, insulin resistance, and dyslipidemia commonly coexist, which sets in motion several dysregulated metabolic pathways inextricably related to oxidative stress and inflammatory processes, ultimately creating a vicious cycle where one process potentiates another 47 , Most notable are the polyol and protein kinase C PKC pathways, which augment oxidative stress and deplete endothelial nitric oxide synthase, respectively, leading to higher endothelin-1 and vascular endothelial growth factor levels.

Endothelial instability and nuclear factor-κB NF-κB —mediated cytokines tumor necrosis factor-α [TNF-α] and interleukin-6 [IL-6] favor an inflammatory response. The hyperglycemic milieu also encourages the accumulation of advanced glycation end products AGEs.

AGEs are a heterogeneous group of nonenzymatically glycated molecules. Upon engaging with their receptors RAGEs , which are found throughout the kidney, they trigger cellular function perturbations, including NF-κB upregulation, which induces a cascade of proinflammatory cytokines TNF-α and IL AGEs reduce the bioavailability of endothelium-derived nitric oxide and increase reactive oxygen species production, which is linked to impaired vasodilatation in diabetes 49 Fig.

Inflammation and fibrosis are major interrelated contributors to DKD progression. Mounting evidence implicates an intricate interaction between the mineralocorticoid receptor MR aldosterone and RAS-related C3 botulinum toxin substrate 1 Rac1 in driving the inflammatory processes that lead to the final common pathway of fibrosis in DKD.

The deleterious effects of MR activation in modulating inflammation and fibrosis were long recognized in animal studies of the heart and provide the therapeutic basis for MR antagonism 50 , Animal studies showed similar benefits of MR blockade on the kidneys 52 and vasculature MR activation occurs in the aldosterone-responsive distal nephron, causing sodium reabsorption and potassium excretion.

Aldosterone secretion is stimulated by RAS activation in response to decreased circulating plasma volume or significant increases in serum potassium levels. While critical for survival in states of low sodium intake, it becomes pathologic in the setting of persistently high sodium intake 54 , as exemplified by Western and many Asian diets.

Inappropriate aldosterone signaling combined with high sodium intake results in hypertension, a direct contributor to glomerular injury and fibrosis.

The MR possesses a binding affinity for cortisol and corticosterone similar to that of aldosterone. These cells typically coexpress 11B-dehydrogenase isoenzyme 2 11B-HSD2 , which neutralizes cortisol and thereby mitigates MR overactivation.

Outside the distal nephron, MRs are expressed on other cell types, including podocytes, fibroblasts, vascular cells, and macrophages; these cells, however, do not uniformly coexpress the steroid-blunting effects of 11B-HSD2, which permits unbridled MR activation MR is upregulated in hyperglycemia, insulin resistance, dyslipidemia, and obesity.

This results in increased gene transcription of profibrotic factors plasminogen activator inhibitor-1 PAI-1 and TGF-β1, connective tissue growth factor, and extracellular matrix proteins, all of which contribute to progressive DKD 56 Fig.

Innate immunity plays a critical role in the pathogenesis of DKD, but a detailed discussion is beyond the scope of this review. Briefly, macrophage infiltration has been identified as one of the hallmarks of DKD, the burden of which is associated with worse disease Hyperglycemia, endothelial cell dysfunction, angiotensin II, AGEs, and oxidized LDL recruit macrophages Macrophage-MR activation polarizes macrophage differentiation toward the M1 phenotype, which promotes inflammation via a cascade of injurious cytokines 59 Figs.

Of note, however, is that MR inhibition favors macrophage switching toward an M2 anti-inflammatory phenotype with demonstrated beneficial effects in CKD The therapeutic effects of MR antagonism in curtailing DKD were first reported in using animal models There are no outcome trials with the steroidal MRAs due to tolerability issues.

However, they have been shown in people with early DKD to reduce albuminuria and blood pressure significantly. Other NS-MRAs, namely, finerenone, esaxerenone, and apararenone, also have demonstrated significant albuminuria reduction and a very low adverse effect profile in advanced DKD 64 , 65 , although esaxerenone is only available in Japan None of these agents have ever been tested in outcome trials.

The timeline of the major therapeutic outcome trials focused on delaying DKD in patients with type 2 diabetes and CKD following the publication of RENAAL and IDNT 73 — MoAs, mechanisms of action.

DKD management has evolved over the last 50 years. However, following the trials that showed a clear benefit of ARBs in slowing DKD progression, nothing was proven to further curtail advancing DKD until the advent of SGLT2 inhibitors.

Many trials using novel targeted therapies were attempted between and , all of which failed to show any benefit in changing the trajectory of DKD Fig. Since the approval of the first SGLT2 inhibitor, dapagliflozin, in January , the field has added other drugs to this class.

Additionally, approval of the NS-MRA finerenone, in July , has further advanced the field from one to three therapies in the tool kit. Questions remain concerning when and how to use these drugs.

Obesity reduces the production of adiponectin in favor of leptin. Gene transcription of inflammatory mediators such as IL-1, IL-6, IL-7, IL-8, and TNF-α is increased, which creates a proinflammatory state and oxidative stress. Profibrotic factors plasminogen activator inhibitor-1 PAI-1 and TGF-β1, connective tissue growth factor, and extracellular matrix proteins are increased, all contributing to progressive diabetic nephropathy.

IL, interleukin; TNF-α, tumor necrosis factor-α; MCP-1, monocyte chemoattractant protein Many trials have established that a reduction in albuminuria is associated with slowed DKD progression and reduced CV event rates 69 — Given the advent of many new therapies over the past decade, we put forth the concept of pillars of therapy, originally adopted by heart failure cardiologists.

The concept is akin to any building structure where no single beam alone can support its standing. Hence, we now have three established, proven therapies that, when used together, will maximally decelerate DKD progression.

This does not imply that those with high albuminuria, formerly called microalbuminuria, do not derive benefit from treatment—in fact, they do, and the benefits are predominantly CV.

However, the slowing of the DKD trajectory in these cohorts was detected in post hoc analyses. This ushered in two additional landmark trials conducted in patients with type 1 diabetes, Reduction of Endpoints in NIDDM with the Angiotensin II Antagonist Losartan RENAAL 20 and Irbesartan Diabetic Nephropathy Trial IDNT 19 , before RAS blockers became formally integrated into the standard of care The introduction of SGLT2 inhibitors in January created a resounding paradigm shift and renewed excitement in improving DKD management.

Although they were originally designed to lower glucose by promoting urinary glucose excretion, it was ultimately appreciated that SGLT2 inhibitors were, in effect, cardiorenal risk—reducing agents The renoprotective benefits of SGLT2 inhibitors were initially gleaned from secondary data analyses of trials with time to major adverse CV events as primary outcomes.

In a meta-analysis of six double-blinded randomized trials of SGLT2 inhibitors in patients with type 2 diabetes, a consistent reduction in hospitalization for heart failure and progression to ESKD was found However, in three landmark trials that followed, where kidney outcomes were the defined primary end points in a high-risk cohort, the benefits of SGLT2 inhibitors when combined with optimized RAS blockade became universally accepted.

The Canagliflozin and Renal Events in Diabetes with Established Nephropathy Clinical Evaluation CREDENCE trial was the first nephropathy outcome trial to specify its primary outcome as ESKD, doubling of creatinine level, or death from renal or CV causes with an SGLT2 inhibitor.

The unequivocal renal benefits prompted early trial termination The Dapagliflozin and Prevention of Adverse Outcomes in Chronic Kidney Disease DAPA-CKD trial, which was published a year later than the CREDENCE trial, randomized a mixed cohort of participants, i. Most recently, The Study of Heart and Kidney Protection With Empagliflozin EMPA-KIDNEY outcome trial adds to the substantial body of evidence underscoring the robust salutary effects of SGLT2 inhibitors on renal outcomes.

Over 6, participants with and without diabetes, with a wide range of GFR declines, from mild to severe, and with normal or high albuminuria, were randomized to empagliflozin versus placebo in addition to standard-of-care therapies.

It should further be noted that there were many people with normal and high albuminuria in this trial. Hence, SGLT2 inhibitors serve as a solid second pillar to slow DKD progression The hazardous sequela of MR activation by aldosterone shifted attention to developing therapies downstream of the RAS target, which ultimately addresses the inflammatory and ensuing profibrotic processes that contribute to DKD progression and heart failure.

MR antagonism is not new. Steroid-based MRAs, including first- and second-generation spironolactone and eplerenone, respectively, continue to be used extensively in symptomatic heart failure patients This has been adopted by the U.

Food and Drug Administration as evidence of benefit and as a guideline by the American Diabetes Association. The exception is when a dual RAS blockade is used. Therefore, MRAs have been generally contraindicated in advanced kidney disease Finerenone, apararenone, esaxerenone, and ocedurenone are members of a new class of NS-MRAs with pharmacologic properties distinct from those of their distant cousins, the steroidal agents.

Finerenone is the only one developed and approved for cardiorenal risk reduction, whereas the others are approved only for blood pressure control, with no outcome data supporting use in DKD. Finerenone demonstrates superior anti-inflammatory and antifibrotic outcomes compared with its steroidal counterparts in preclinical studies 92 — Unlike the steroidal MRAs, the NS-MRAs achieve balanced tissue distribution between the heart and kidney rather than affecting the kidney alone Finerenone was studied in two complementary phase 3 randomized, double-blinded, placebo-controlled clinical trials that included over 13, participants with type 2 diabetes optimized on maximally tolerated RAS blockade before randomization to the NS-MRA or placebo 96 , These trials were developed with the same protocol but different inclusion criteria, which allowed for an individual pooled patient analysis in the Finerenone in Chronic Kidney Disease and Type 2 Diabetes: Combined FIDELIO-DKD and FIGARO-DKD Trial Programme Analysis FIDELITY Despite the similar frequency of adverse effects in both groups, the incidence of trial discontinuation related to hyperkalemia was slightly higher with finerenone than with placebo 2.

The frequency of reported adverse effects was similar to that for placebo, and the incidence of trial discontinuation related to hyperkalemia was slightly higher with finerenone than with placebo 1.

The FIDELITY individual pooled patient analysis included 13, patients with type 2 diabetes and a wide range of CKD stages, from 1 to 4, and high to very high albuminuria.

This is a very important finding given the notably higher risk of heart failure and premature death associated with having both diabetes and CKD 4 , 99 , Studies of finerenone in type 1 diabetes are currently underway.

The incidence of clinically important adverse effects related to hyperkalemia was slightly higher for finerenone, with 1.

However, unlike many other trials, all participants were required to be on maximally tolerated RAS blockade. Differences in hyperkalemia risk between the NS-MRA finerenone and its steroidal counterpart, spironolactone, are exemplified by a comparative outcome of a subgroup with resistant hypertension from the FIDELITY analysis and the Spironolactone With Patiromer in the Treatment of Resistant Hypertension in Chronic Kidney Disease AMBER trial In the FIDELITY study, the incidence of hyperkalemia was GLP-1 RAs are recommended for patients with DKD who have not met their glycemic targets despite optimization with metformin and SGLT2 inhibitors Post hoc analyses of these CVOT, like analyses of SGLT2 inhibitors, also demonstrated possible benefits in delaying DKD progression Additionally, a large analysis of more than 12, participants pooled from the earliest trials of GLP-1 RAs, Trial to Evaluate Cardiovascular and Other Long-term Outcomes With Semaglutide in Subjects With Type 2 Diabetes SUSTAIN-6 and Liraglutide Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results LEADER , evaluated changes in albuminuria, rate of annual eGFR change, and time to persistent eGFR declines.

Semaglutide and liraglutide were associated with significant slowing of annual eGFR decline, 0. This trial enrolled 4, patients with known type 2 diabetes and either current CKD eGFR Notably, there was a higher prevalence of CKD in the trial cohort than in the seven CVOT studies completed to date.

Additionally, Since Abdominal obesity is independently associated with albuminuria despite normoglycemia and normotension and addresses an important subgroup of patients who have obesity-related glomerulopathy and who, by conventional screening, may be classified as metabolically healthy yet are still at risk for developing ESKD This is not surprising given the inflammatory milieu engendered by excess adiposity Fig.

FLOW A Research Study to See How Semaglutide Works Compared to Placebo in People With Type 2 Diabetes and Chronic Kidney Disease is an ongoing randomized placebo-controlled trial that assesses the efficacy of semaglutide in type 2 diabetes and CKD and the first designed with primary renal end points The renoprotective properties associated with GLP-1 RAs reviewed here were drawn from post hoc analyses of major CVOT, the evidence of which has been compelling, nonetheless.

DKD management should start with maximally dosed RAS blockade; this is based on the doses used in the original RAS blocking trials, wherein dose reduction to avoid hyperkalemia resulted in markedly reduced protection against DKD decline.

Each drug class that provided improved outcomes was coupled with RAS blockade and independently showed benefits on kidney and heart outcomes. These findings provide impetus to adopt a pillared approach for reducing cardiorenal events similar to how heart failure cardiologists have approached heart failure management.

Notably, there has never been a trial that evaluated the simultaneous use of all four agents in heart failure or that compared different drug combinations against each other.

Each trial was assessed on its merits within each drug class and then combined in retrospect to provide the best results. We currently have three agents with additive effects on albuminuria and heart failure outcomes when combined individually with RAS blockade.

An adequately statistically powered study that evaluates the three, or possibly soon-to-be four, different agents together would require well over , participants; therefore, extrapolation, which the cardiologists have done successfully, uses all three agents together over a short time.

Practice guidelines articulate that clinicians should start first by titrating to maximally tolerated RAS blockade before introducing these medications SGLT2 inhibitors, NS-MRAs, and GLP-1 RAs , as was done in pivotal clinical trials Initial changes in eGFR and associated long-term amelioration of renal function decline have also been reported with SGLT2 inhibitors and finerenone, albeit to a lesser degree than with RAS blockers 96 , , — The manifold pathophysiological mechanisms involved in end-organ damage argue for a pillared approach with targeted therapies that have distinct pharmacodynamic actions An elegant study that used an animal model of preclinical hypertension—induced cardiorenal disease with a low-dose combination therapy of finerenone and empagliflozin revealed additive cardiorenal benefit above that of the respective dose-dependent monotherapy, as measured by reductions in blood pressure, proteinuria, cardiac fibrosis, vasculopathy, and mortality These findings further argue strongly for distinct pharmacodynamic actions that counteract the manifold pathophysiological mechanisms involved in end-organ damage Fig.

Acute changes in GFR slopes with three distinct classes of drugs with unique mechanisms that slow kidney disease progression associated diabetes. B : Rate of eGFR decline between finerenone and placebo. Despite equivalent GFR at baseline, the finerenone group shows stabilization of slowed GFR declines 96 and higher GFR declines at 4 months, which are associated with better long-term outcomes.

C : Initial decline in GFR decline in the empagliflozin group compared with placebo at month 4 EMPA-REG Outcome, BI Empagliflozin Cardiovascular Outcome Event Trial in Type 2 Diabetes Mellitus Patients; LS, least squares. Additionally, in a small open-label randomized crossover clinical study, the efficacy and safety of dapagliflozin and a low dose of the steroidal MRA eplerenone were evaluated in a cohort of patients with CKD.

The combination of the two drugs was associated with an additive effect on albuminuria reduction compared with the use of either drug alone. Importantly, the incidence of hyperkalemia was significantly less in the combination group than in the group that received eplerenone alone This is consistent with data from the larger FIDELIO-DKD trial that demonstrated greater protection from hyperkalemia when an SGLT2 inhibitor was combined with finerenone The use of combination therapies with NS-MRAs and SGLT2 inhibitors was further explored in the FIDELITY subgroup analysis, which revealed that, compared with placebo, cardiorenal benefits of finerenone were appreciably higher irrespective of concomitant GLP-1RA or SGLT2 inhibitor use at baseline or anytime during the trial.

An important caveat is that GLP-1 RA and SGLT2 inhibitor users comprised 6. More importantly, there was no sign that drug coadministration with finerenone increased any risk of kidney injury When combined with finerenone, SGLT2 inhibitors reduced hyperkalemic events compared with levels found in nonusers 8.

The patients with transient exposure to hyperglycemia develop diabetic complications, including DKD, even after normalization of their blood glucose.

Temporary hyperglycemia causes advanced glycation end product AGE accumulations and epigenetic changes as metabolic memory. The drugs that improve metabolic memory are awaited, and AGE inhibitors and histone modification inhibitors are the focus of clinical and basic research. In addition, incretin-related drugs showed a renoprotective ability in many clinical trials, and these trials with renal outcome as their primary endpoint are currently ongoing.

Hypoxia-inducible factor prolyl hydroxylase inhibitors recently approved for renal anemia may be renoprotective since they improve tubulointerstitial hypoxia. Thus, following SGLT2 inhibitor, numerous novel drugs could be utilized in treating DKD.

Future studies are expected to provide new insights. Keywords : Diabetic nephropathies ; Epigenomics ; Glycation end products, advanced ; Hypoxia-inducible factor 1 ; NF-E2-related factor 2 ; Sodium-glucose transporter 2. The concept of diabetic kidney disease and diabetic nephropathy.

GFR, glomerular filtration rate. The time course of current treatments. The time course of future treatments.

Received date: Advancfments 21, ; Nephropafhy date: March 14, ; Nelhropathy date: March Electrolyte balance mechanisms, Citation: Ti M, Xue L, Yin Q, Shao S, Diqbetic J, Diabetic nephropathy research advancements al. J Diabetic nephropathy research advancements Exp Nephrol doi: Copyright: © Ti M, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Aim: This article focuses on the genetic mechanisms, with an aim to provide certain theoretical and experimental foundation for the future diagnosis and treatment of diabetic nephropathy.

Author: Gagor

4 thoughts on “Diabetic nephropathy research advancements

  1. Ich meine, dass Sie sich irren. Ich kann die Position verteidigen. Schreiben Sie mir in PM, wir werden besprechen.

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com