ArticlePDF Available

A Perspective on the Impact of Advanced Glycation End Products in the Progression of Diabetic Nephropathy

Authors:

Abstract

In 2007, diabetes affected around 244 million people across the globe. The number of diabetics worldwide is projected to reach 370 million by 2030. With diabetes incidence reaching epidemic proportions globally, diabetic nephropathy (DN) has emerged as one of the most difficult health conditions. Although therapeutic approaches such as rigorous blood glucose and blood pressure management are successful in preventing DN, they are far from ideal, and the number of diabetic patients with endstage renal disease continues to grow. As a result, a unique treatment approach for DN should be devised. There is mounting evidence that advanced glycation end products (AGEs), senescent macro protein derivatives generated at an accelerated pace in DN, contribute to DN by generating oxidative stress. The purpose of
Current Protein & Peptide Science
ISSN: 1389-2037
eISSN: 1875-5550
Impact
Factor:
3.118
SCIENCE
BENTHAM
Current
Science
Protein & Peptide
The international
journal for timely
in-depth reviews in
Protein & Peptide
Science
Send Orders for Reprints to reprints@benthamscience.net
2
Current Protein and Peptide Science, 2023, 24, 2-6
PERSPECTIVE
A Perspective on the Impact of Advanced Glycation End Products in the
Progression of Diabetic Nephropathy
1875-5550/23 $65.00+.00 © 2023 Bentham Science Publishers
Afreen Khanam1, Saheem Ahmad2, and Arbab Husain1,*
1Department of Biotechnology & Life Science, Institute of Biomedical Education & Research, Mangalayatan University,
Aligarh, Uttar Pradesh, India; 2Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Uni-
versity of Hail, Hail-2440, Saudi Arabia
Abstract: In 2007, diabetes affected around 244 million people across the globe. The number of dia-
betics worldwide is projected to reach 370 million by 2030. With diabetes incidence reaching epidemic
proportions globally, diabetic nephropathy (DN) has emerged as one of the most difficult health condi-
tions. Although therapeutic approaches such as rigorous blood glucose and blood pressure management
are successful in preventing DN, they are far from ideal, and the number of diabetic patients with end-
stage renal disease continues to grow. As a result, a unique treatment approach for DN should be de-
vised. There is mounting evidence that advanced glycation end products (AGEs), senescent macro pro-
tein derivatives generated at an accelerated pace in DN, contribute to DN by generating oxidative
stress. The purpose of this article is to discuss the pathophysiological significance of AGEs and their
receptor in DN.
A R T I C L E H I S T O R Y
Received: June 09, 2022
Revised: August 26, 2022
Accepted: September 06, 2022
DOI:
10.2174/1389203724666221108120715
Keywords: Diabetes mellitus, diabetic nephropathy, glycation, AGEs, RAGE, renal disease.
1. INTRODUCTION
Diabetes nephropathy is the most common cause of end-
stage renal disease globally and may contribute to the disa-
bility and high death rate associated with diabetes. Accord-
ing to the World Health Organization, the worldwide diabe-
tes population will reach 370 million by 2030 [1]. Current
treatment alternatives, however, are far from adequate, and
the results of rigorous therapy on DN are inadequate. Indeed,
extensive management of blood glucose and/or blood pres-
sure is often difficult to maintain in diabetic patients and
may raise the risk of hypoglycemia and/or hypotension, and
the number of diabetic patients with end-stage renal failure
continues to rise in developed nations. As a result, the devel-
opment of innovative treatment techniques that are specifi-
cally targeted at DN is essential [2].
Throughout their lifespan, proteins and polypeptides are
frequently exposed to a variety of post-translational changes,
including phosphorylation, glycation, deamination, ubiquiti-
nation, and sumoylation [3]. These post-translational chang-
es may aid proteins in folding and sorting into diverse cellu-
lar compartments, trigger intracellular signaling, confer sta-
bility under stress circumstances, and regulate their intracel-
lular levels via degradation and control [4]. Proteins are also
subject to post-translational alterations that may affect their
*Address correspondence to this author at the Department of Biotechnology
& Life Sciences, Institute of Biomedical Education & Research, Man-
galayatan University, Aligarh, Uttar Pradesh, India;
E-mail: arbabhusain1@gmail.com
structure, function, and half-life due to natural aging and
pathological diseases such as diabetes. Glycation is one such
post-translational modification. Glycation of client proteins
results in crosslinking and aggregation, which may have a
detrimental effect on proteins' structural and functional char-
acteristics [5]. Along with altering the physicochemical
characteristics of the affected proteins, glycation causes ab-
normal cellular signaling, transcription factor activation, and
alterations in gene expression patterns. The end products
formed as a result of glycation are referred to as AGEs [6].
Even though glycation occurs at a slower rate throughout
life, it accelerates in clinical conditions such as diabetes and
is implicated in the pathophysiology of several diabetes
complications and other age-related disorders such as cata-
racts, nephropathy, cardiovascular complications, and Alz-
heimer's disease [7]. In individuals with T2DM, tissue AGEs
are highly correlated with early kidney, ocular, and nerve
damage [8].
2. AGEs AND DN
Both clinical and experimental evidence suggests that
AGEs are involved in the etiology of diabetic complications,
particularly DN, in type 1 and type 2 diabetes [9]. DN is the
most prevalent microvascular chronic complication, occur-
ring in 20%–40% of type 1 and type 2 diabetes mellitus
(T2DM) patients, respectively. Around one-third of all new
instances of end-stage renal disease (ESRD) are caused by
DN [10]. Affected glomerular capillaries result in a gradual
loss in glomerular filtration rate, which is followed by many
A Perspective on the Impact of Advanced Glycation End Products Current Protein and Peptide Science, 2023, Vol. 24, No. 1 3
stages of proteinuria, including microalbuminuria, macroal-
buminuria, and overt proteinuria, and ultimately resulting in
ESRD necessitating renal replacement. Hyperglycemia and
hypertension, which are also characteristics of T2DM, exac-
erbate the progression to ESRD [11]. Renal disease is char-
acterized by both hemodynamic (hyperfiltration) and struc-
tural abnormalities in diabetic patients. Significant glomeru-
lar alterations include basement membrane thickening,
mesangial enlargement and hypertrophy, and podocyte loss
[12]. The tubulointerstitial compartment undergoes signifi-
cant alterations throughout DN, including tubular basement
membrane thickening, tubular atrophy, interstitial fibrosis,
and arteriosclerosis [13]. The crucial involvement of AGEs
in the etiology of renal damage was shown in work in nondi-
abetic rats, where AGEs generated proteinuria and degenera-
tive alterations similar to those seen in DN. When type 1
diabetes patients progress from normal renal function to
ESRD, blood levels of fluorescent-CML AGEs rapidly rise.
In contrast, another research discovered a correlation be-
tween CML and the severity of nephropathy in type 1 diabe-
tes individuals. There is a considerable rise in CML and hy-
droimidazolone-AGEs in T2DM individuals. Serum CML-
human proteins were significantly higher in individuals with
proteinuria, and a rise in circulating AGEs peptides was as-
sociated with the degree of renal impairment [14]. The sever-
ity of DN is related to the number of AGEs formed and the
number of receptors for advanced glycation end products
(RAGE) expressed in the glomerular and tubulointerstitial
compartments. Accumulation of AGEs in the kidney may
contribute to the gradual change of renal architecture and
loss of renal function in humans and rodents through many
processes, such as the cross-linking of matrix proteins and
activation of downstream signaling. The development of
AGEs on extracellular matrix proteins modifies both matrix-
matrix and cell-matrix interactions and is implicated in dia-
betic glomerulosclerosis. AGEs promote apoptotic cell death
and VEGF expression in human mesangial cells cultivated in
vitro, much like in pericytes, the retinal analogue of mesan-
gial cells. As mesangial cells occupy a central anatomical
position in the glomerulus and play a crucial role in main-
taining the structure and function of glomerular capillary
tufts, the AGEs-induced apoptosis and dysfunction of
mesangial cells may contribute in part to glomerular hyper-
filtration, an early renal dysfunction in diabetes [15]. Addi-
tionally, the course of DN is marked by a reduction in the
glomerular filtration rate (GFR). AGEs increase the synthe-
sis of asymmetric dimethylarginine (ADMA), an endogenous
inhibitor of nitric oxide (NO) synthase. Plasma ADMA lev-
els are favorably connected with serum AGEs levels and
were adversely correlated with endothelial function evaluat-
ed by flow-mediated vasodilatation. In addition, AGEs low-
ered the messenger RNA (mRNA) level of dimethylarginine
dimethylaminohydrolase (DDAH)-II, an enzyme for ADMA
degradation, decreased its overall enzymatic activity, and
increased ADMA, all of which were prevented by the antiox-
idant N-acetylcysteine. This implies that the AGE-RAGE-
mediated ROS production might be implicated in endothelial
dysfunction in diabetic ESRD patients partially by boosting
the ADMA formation via reduction of DDAH activity in
ECs [16]. The inhibition of renal NO generation by AGEs is
thought to be a factor in the reduced GFR [17].
3. AGEs AFFECT ECM METABOLISM AND CAUSE
DN
DN is distinguished by the accumulation of ECM in the
mesangium, tubular interstitium, and glomerular basement
membrane (GBM) [18]. AGEs disrupt the equilibrium of
ECM component production and breakdown, notably colla-
gen. Collagen is one of the most thoroughly researched pro-
teins for glycation modification. AGEs development occurs
as a result of intra and intermolecular crosslinks in collagen,
resulting in major structural alterations such as changed
packing density and surface charge, all of which result in
lower solubility [19]. Glycated collagen impairs its ability to
self-assemble and interact with sub-ECM components. Re-
duced flexibility of arterial vessel walls and capillaries in the
glomeruli results from impaired intermolecular interactions
within collagen fibers and other components of basement
membranes [20]. Glycation of collagen has long been linked
to the progression of diabetes-related secondary complica-
tions. How precisely such random glycations arise in dam-
aged tissues is currently poorly understood. Because most
fibrillar collagens have a sluggish turnover rate, they are
more sensitive to accumulating time-dependent glycations
and advanced glycation end products. It is hypothesized that
the latter contain cross-links that stiffen host tissues. Howev-
er, diabetic animal models had weaker tendons with de-
creased rigidity. Using targeted mass spectrometry (MS),
researchers discovered higher partial fructosyl-hydroxylysine
glycations at each of the helical domain cross-linking sites of
type I collagen in tissues from a mouse model of diabetes.
None of the other collagen lysine residues were glycated. In
the tendons of mice, type I collagen is intermolecularly
cross-linked by acid-labile aldimine linkages generated by
the addition of telopeptide lysine aldehydes to hydroxylysine
residues at positions α1(I)Lys87, α1(I)Lys930, α2(I)Lys87,
and α2(I)Lys933 of the triple helix. These findings indicate
that site-specific glycation of these specific lysines may sig-
nificantly impair normal lysyl oxidase–controlled cross-
linking in diabetic tendons, suggesting that such N-linked
glycations may alter the content and/or placement of mature
cross-links thereby having the potential to modify tissue ma-
terial properties [21]. Aside from influencing collagen me-
tabolism, AGEs impact the interactions of ECM components.
Glycation may disturb intra-ECM and cell-matrix interac-
tions, leading to decreased cell proliferation, altered cellular
adhesion, and loss of the epithelial phenotype [5]. The modi-
fication of numerous ECM proteins with AGEs hinders their
breakdown by matrix metalloproteinases, which contributes
to basement membrane thickening and mesangial expansion
[22].
Due to charge-charge repulsion, the ECM of podocytes
and renal capillaries is negatively charged, preventing serum
albumin leakage into the glomerular filtrate. Glycation and
buildup of AGEs in the ECM components of renal capillar-
ies, on the other hand, modify the electrostatic interactions
that result in altered vascular permeability to albumin [23].
Accumulation of AGEs in ECM components may trap plas-
ma proteins, lipoproteins, and immunoglobulins, impairing
normal renal function balance and aggravating glomerulo-
sclerosis. Glycated collagen from the basement membrane
promotes platelet aggregation. The detrimental effects of
AGE buildup on ECM components have also been shown in
4 Current Protein and Peptide Science, 2023, Vol. 24, No. 1 Khanam et al.
investigations using AGE breakers. ALT-711 and N-
phenacylthiazolium bromide cleavage of AGE-induced
crosslinks increased collagen solubility and decreased matrix
formation in the kidney [24]. In podocytes, tubular cells, and
mesangial cells, AGEs promote the expression of transform-
ing growth factor-β (TGF-β ). TGF-β is a pro-fibrotic factor
that increases type IV collagen, laminin, and fibronectin syn-
thesis [25]. Additionally, AGEs stimulate the production of
platelet-derived growth factor, which is pro-fibrotic. Apart
from hyperglycemia, glycoxidation and lipoxidation prod-
ucts, such as CML, pentosidine, and malondialdehyde-
lysine, also contribute to the formation of AGEs [26]. Col-
lectively, AGEs change the metabolism and expression of
ECM components, resulting in an advanced nephropathy
phenotype.
4. ROLE OF RAGE AND OXIDATIVE STRESS IN DN
There is mounting evidence that the AGE-RAGE axis
plays a role in DN development. Among the different kinds
of AGEs receptors, RAGE is a signal-transducing receptor
for AGEs that may play a role in mediating the inflammatory
responses induced by AGEs [27]. In diabetic individuals
with nephropathy, RAGE expression is increased in podo-
cytes and mesangial cells. Numerous animal investigations
have also shown that RAGE plays a critical role in the estab-
lishment and progression of DN [28]. Indeed, diabetic mice
overexpressing RAGE exhibit increasing glomerulosclerosis
and renal failure compared to diabetic littermates without the
RAGE transgene. Additionally, it was reported that activat-
ing RAGE in podocytes might result in increased VEGF
expression and increased attraction/activation of inflammato-
ry cells in diabetic glomeruli, resulting in albuminuria and
glomerulosclerosis [29]. Furthermore, it has been reported
that diabetic mice induced with dB/dB or streptozotocin de-
velop renal changes similar to those seen in human DN, in-
cluding glomerular hypertrophy, glomerular basement mem-
brane thickening, mesangial matrix expansion, connective
tissue growth factor (CTGF) overexpression, and NFκ-B
activation, all of which are prevented by the administration
of a neutralizing antibody raised against RAGE. Further-
more, the AGE-RAGE interaction may result in chronic acti-
vation of NFκB due to higher amounts of de novo produced
NFκBp65 overcoming endogenous negative feedback sys-
tems, thereby contributing to the persistence of diabetic kid-
ney damage [15]. RAGE interaction with AGEs results in the
formation of oxidative stress, which contributes to DN. AG-
Es bind with their particular cell-surface receptor, RAGE,
initiating a signaling cascade that activates the transcription
factor nuclear factor-kappa B (NF-κB), resulting in an in-
crease in the production of inflammatory cytokines such as
TNF-α. The ensuing rise in inflammation leads to cellular
dysfunction and tissue destruction. Furthermore, AGEs in-
hibit intracellular detoxification mechanisms, which result in
increased oxidative stress. This AGE-RAGE axis, through
activating NADPH oxidases and/or by other similar mecha-
nisms, also stimulates the generation of reactive oxygen spe-
cies (ROS), which exacerbate oxidative damage to cells [30].
Indeed, ROS are cytotoxic to renal cells and promote in-
flammatory and fibrogenic responses in diabetic kidneys
[31]. The generation of ROS by AGE-RAGE induces the
synthesis of pro-sclerotic growth factors such as TGFβ and
CTGF in mesangial and renal tubulointerstitial cells through
the mitogen-activated protein kinase (MAPK), NFκ-B,
and/or PKC pathways.
Recent research has demonstrated the significance of Di-
aphanous1 in mouse diabetic kidney disease (DKD). Similar
to results involving RAGE expression in DKD, it was dis-
covered that Diaphanous1 was expressed in the tubulo inter-
stitium and podocytes of the human and mouse diabetic kid-
ney. Streptozotocin produced T1D-like symptoms in Diaph-
anous1 deficient and wild-type mice. After 6 months, diabet-
ic Diaphanous1 null mice exhibited considerably less
mesangial sclerosis, podocyte effacement, glomerular base-
ment thickening, and urinary albumin excretion than diabetic
wild-type control animals. Examining the renal cortex of
diabetic mice indicated that deletion of Diaphanous1 greatly
decreased the expression of genes associated with fibrosis
and inflammation [32]. Additionally, it was recently discov-
ered that inhibiting NADPH oxidase with apocynin protects
the kidneys against AGE-induced damage in experimental
DN through a PKC-dependent pathway. Thus, inhibiting
NADPH oxidase-derived reactive oxygen species (ROS)
production provoked by the AGE-RAGE system may be a
potential therapeutic target for diabetic patients with
nephropathy [33, 34].
CONCLUSION
With the enormity of diabetes incidence, the impact of
diabetes on human health, and the relevance of renal func-
tion, it is critical to have effective diabetes prevention strate-
gies. The scarcity of inexpensive and effective renal treat-
ment for those with chronic kidney disease adds to the global
healthcare burden. The body of data suggests that AGEs play
a crucial role in the pathogenesis of DN. As a consequence,
evaluating the effectiveness and safety of anti-AGE drugs
such as AGE blockers/crosslink breakers, ACE inhibitors,
and sRAGE inhibitors is the preferred preventative medica-
tion for DN.
LIST OF ABBREVIATIONS
AGEs = Advanced Glycation End Products
DN = Diabetic Nephropathy
T2DM = Type 2 Diabetes Mellitus
RAGE = Receptor for Advanced Glycation End Products
ESRD = End Stage Renal Disease
CML = Carboxy Methyl Lysine
GBM = Glomerular Basement Membrane
ECM = Extra Cellular Matrix
ADMA = Asymmetric Dimethylarginine
DDAH = Dimethylarginine Dimethylaminohydrolase
TGF-β = Transforming Growth Factor-β
CTGF = Connective Tissue Growth Factor
MAPK = Mitogen-Activated Protein Kinase
A Perspective on the Impact of Advanced Glycation End Products Current Protein and Peptide Science, 2023, Vol. 24, No. 1 5
CONSENT FOR PUBLICATION
Not applicable.
FUNDING
None.
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or
otherwise.
ACKNOWLEDGEMENTS
The author would like to acknowledge Mangalayatan
University, Aligarh, and the University of Hail, Hail, Saudi
Arabia.
REFERENCES
[1] Bikbov, B.; Purcell, C.A.; Levey, A.S.; Smith, M.; Abdoli, A.;
Abebe, M.; Adebayo, O.M.; Afarideh, M.; Agarwal, S.K.; Agude-
lo-Botero, M.; Ahmadian, E.; Al-Aly, Z.; Alipour, V.; Almasi-
Hashiani, A.; Al-Raddadi, R.M.; Alvis-Guzman, N.; Amini, S.;
Andrei, T.; Andrei, C.L.; Andualem, Z.; Anjomshoa, M.; Arabloo,
J.; Ashagre, A.F.; Asmelash, D.; Ataro, Z.; Atout, M.M.W.;
Ayanore, M.A.; Badawi, A.; Bakhtiari, A.; Ballew, S.H.; Balouchi,
A.; Banach, M.; Barquera, S.; Basu, S.; Bayih, M.T.; Bedi, N.; Bel-
lo, A.K.; Bensenor, I.M.; Bijani, A.; Boloor, A.; Borzì, A.M.;
Cámera, L.A.; Carrero, J.J.; Carvalho, F.; Castro, F.; Catalá-López,
F.; Chang, A.R.; Chin, K.L.; Chung, S.-C.; Cirillo, M.; Cousin, E.;
Dandona, L.; Dandona, R.; Daryani, A.; Das Gupta, R.; Demeke,
F.M.; Demoz, G.T.; Desta, D.M.; Do, H.P.; Duncan, B.B.; Eftek-
hari, A.; Esteghamati, A.; Fatima, S.S.; Fernandes, J.C.; Fernandes,
E.; Fischer, F.; Freitas, M.; Gad, M.M.; Gebremeskel, G.G.; Gebre-
sillassie, B.M.; Geta, B.; Ghafourifard, M.; Ghajar, A.; Ghith, N.;
Gill, P.S.; Ginawi, I.A.; Gupta, R.; Hafezi-Nejad, N.; Haj-Mirzaian,
A.; Haj-Mirzaian, A.; Hariyani, N.; Hasan, M.; Hasankhani, M.;
Hasanzadeh, A.; Hassen, H.Y.; Hay, S.I.; Heidari, B.; Herteliu, C.;
Hoang, C.L.; Hosseini, M.; Hostiuc, M.; Irvani, S.S.N.; Islam,
S.M.S.; Jafari Balalami, N.; James, S.L.; Jassal, S.K.; Jha, V.; Jo-
nas, J.B.; Joukar, F.; Jozwiak, J.J.; Kabir, A.; Kahsay, A.; Ka-
saeian, A.; Kassa, T.D.; Kassaye, H.G.; Khader, Y.S.; Khalilov, R.;
Khan, E.A.; Khan, M.S.; Khang, Y.-H.; Kisa, A.; Kovesdy, C.P.;
Kuate Defo, B.; Kumar, G.A.; Larsson, A.O.; Lim, L.-L.; Lopez,
A.D.; Lotufo, P.A.; Majeed, A.; Malekzadeh, R.; März, W.; Masa-
ka, A.; Meheretu, H.A.A.; Miazgowski, T.; Mirica, A.; Mir-
rakhimov, E.M.; Mithra, P.; Moazen, B.; Mohammad, D.K.; Mo-
hammadpourhodki, R.; Mohammed, S.; Mokdad, A.H.; Morales,
L.; Moreno Velasquez, I.; Mousavi, S.M.; Mukhopadhyay, S.; Na-
chega, J.B.; Nadkarni, G.N.; Nansseu, J.R.; Natarajan, G.; Nazari,
J.; Neal, B.; Negoi, R.I.; Nguyen, C.T.; Nikbakhsh, R.; Noubiap,
J.J.; Nowak, C.; Olagunju, A.T.; Ortiz, A.; Owolabi, M.O.; Palladi-
no, R.; Pathak, M.; Poustchi, H.; Prakash, S.; Prasad, N.; Rafiei, A.;
Raju, S.B.; Ramezanzadeh, K.; Rawaf, S.; Rawaf, D.L.; Rawal, L.;
Reiner, R.C. Jr.; Rezapour, A.; Ribeiro, D.C.; Roever, L.; Rothen-
bacher, D.; Rwegerera, G.M.; Saadatagah, S.; Safari, S.; Sahle,
B.W.; Salem, H.; Sanabria, J.; Santos, I.S.; Sarveazad, A.;
Sawhney, M.; Schaeffner, E.; Schmidt, M.I.; Schutte, A.E.;
Sepanlou, S.G.; Shaikh, M.A.; Sharafi, Z.; Sharif, M.; Sharifi, A.;
Silva, D.A.S.; Singh, J.A.; Singh, N.P.; Sisay, M.M.M.; Soheili, A.;
Sutradhar, I.; Teklehaimanot, B.F.; Tesfay, B.; Teshome, G.F.;
Thakur, J.S.; Tonelli, M.; Tran, K.B.; Tran, B.X.; Tran Ngoc, C.;
Ullah, I.; Valdez, P.R.; Varughese, S.; Vos, T.; Vu, L.G.; Waheed,
Y.; Werdecker, A.; Wolde, H.F.; Wondmieneh, A.B.; Wulf Han-
son, S.; Yamada, T.; Yeshaw, Y.; Yonemoto, N.; Yusefzadeh, H.;
Zaidi, Z.; Zaki, L.; Zaman, S.B.; Zamora, N.; Zarghi, A.; Zewdie,
K.A.; Ärnlöv, J.; Coresh, J.; Perico, N.; Remuzzi, G.; Murray,
C.J.L.; Vos, T. Global, regional, and national burden of chronic
kidney disease, 19902017: A systematic analysis for the Global
Burden of Disease Study 2017. Lancet, 2020, 395(10225), 709-
733.
http://dx.doi.org/10.1016/S0140-6736(20)30045-3 PMID:
32061315
[2] Husain, A.; Farooqui, A.; Khanam, A.; Sharma, S.; Mahfooz, S.;
Shamim, A.; Akhter, F.; Alatar, A.A.; Faisal, M.; Ahmad, S. Physi-
cochemical characterization of C-phycocyanin from Plectonema
sp. and elucidation of its bioactive potential through in silico ap-
proach. Cell. Mol. Biol., 2022, 67(4), 68-82.
http://dx.doi.org/10.14715/cmb/2021.67.4.8 PMID: 35809301
[3] Gupta, R.; Sahu, M.; Srivastava, D.; Tiwari, S.; Ambasta, R.K.;
Kumar, P. Post-translational modifications: Regulators of neuro-
degenerative proteinopathies. Ageing Res. Rev., 2021, 68, 101336.
http://dx.doi.org/10.1016/j.arr.2021.101336 PMID: 33775891
[4] Niforou, K.; Cheimonidou, C.; Trougakos, I.P. Molecular chaper-
ones and proteostasis regulation during redox imbalance. Redox
Biol., 2014, 2, 323-332.
http://dx.doi.org/10.1016/j.redox.2014.01.017 PMID: 24563850
[5] Kumar Pasupulati, A.; Chitra, P.S.; Reddy, G.B. Advanced gly-
cation end products mediated cellular and molecular events in the
pathology of diabetic nephropathy. Biomol. Concepts, 2016, 7(5-6),
293-309.
http://dx.doi.org/10.1515/bmc-2016-0021 PMID: 27816946
[6] Briceno Noriega, D.; Zenker, H.E.; Croes, C.A.; Ewaz, A.; Ruine-
mans-Koerts, J.; Savelkoul, H.F.J.; van Neerven, R.J.J.; Teodoro-
wicz, M. Receptor mediated effects of advanced glycation end
products (AGEs) on innate and adaptative immunity: Relevance for
food allergy. Nutrients, 2022, 14(2), 371.
http://dx.doi.org/10.3390/nu14020371 PMID: 35057553
[7] Rowan, S.; Bejarano, E.; Taylor, A. Mechanistic targeting of ad-
vanced glycation end-products in age-related diseases. Biochim.
Biophys. Acta Mol. Basis Dis., 2018, 1864(12), 3631-3643.
http://dx.doi.org/10.1016/j.bbadis.2018.08.036 PMID: 30279139
[8] Moldogazieva, N.T.; Mokhosoev, I.M.; Mel’nikova, T.I.; Porozov,
Y.B.; Terentiev, A.A. Oxidative stress and advanced lipoxidation
and glycation end products (ALEs and AGEs) in aging and age-
related diseases. Oxid. Med. Cell. Longev., 2019, 2019, 1-14.
http://dx.doi.org/10.1155/2019/3085756 PMID: 31485289
[9] Papadopoulou-Marketou, N.; Chrousos, G.P.; Kanaka-Gantenbein,
C. Diabetic nephropathy in type 1 diabetes: A review of early natu-
ral history, pathogenesis, and diagnosis. Diabetes Metab. Res. Rev.,
2017, 33(2), e2841.
http://dx.doi.org/10.1002/dmrr.2841 PMID: 27457509
[10] Gheith, O.; Farouk, N.; Nampoory, N.; Halim, M.A.; Al-Otaibi, T.
Diabetic kidney disease: Worldwide difference of prevalence and
risk factors. J. Nephropharmacol., 2015, 5(1), 49-56.
PMID: 28197499
[11] Anders, H.J.; Huber, T.B.; Isermann, B.; Schiffer, M. CKD in
diabetes: Diabetic kidney disease versus nondiabetic kidney dis-
ease. Nat. Rev. Nephrol., 2018, 14(6), 361-377.
http://dx.doi.org/10.1038/s41581-018-0001-y PMID: 29654297
[12] Amann, K.; Benz, K. Structural renal changes in obesity and diabe-
tes. Semin. Nephrol., 2013, 33(1), 23-33.
http://dx.doi.org/10.1016/j.semnephrol.2012.12.003 PMID:
23374891
[13] Habib, S.L. Kidney atrophy vs. hypertrophy in diabetes: Which
cells are involved. Cell Cycle, 2018, 17(14), 1683-1687.
http://dx.doi.org/10.1080/15384101.2018.1496744 PMID:
29995580
[14] Tan, A.L.Y.; Forbes, J.M.; Cooper, M.E. AGE, RAGE, and ROS in
diabetic nephropathy. Semin. Nephrol., 2007, 27(2), 130-143.
http://dx.doi.org/10.1016/j.semnephrol.2007.01.006 PMID:
17418682
[15] Yamagishi, S.; Nakamura, N.; Suematsu, M.; Kaseda, K.; Matsui,
T. Advanced glycation end products: A molecular target for vascu-
lar complications in diabetes. Mol. Med., 2015, 21(S1)(Suppl. 1),
S32-S40.
http://dx.doi.org/10.2119/molmed.2015.00067 PMID: 26605646
[16] Ando, R.; Ueda, S.; Yamagishi, S.; Miyazaki, H.; Kaida, Y.; Kaifu,
K.; Yokoro, M.; Nakayama, Y.; Obara, N.; Fukami, K.; Takeuchi,
M.; Okuda, S. Involvement of advanced glycation end product-
induced asymmetric dimethylarginine generation in endothelial
dysfunction. Diab. Vasc. Dis. Res., 2013, 10(5), 436-441.
http://dx.doi.org/10.1177/1479164113486662 PMID: 23766377
6 Current Protein and Peptide Science, 2023, Vol. 24, No. 1 Khanam et al.
[17] Khanam, A.; Ahmad, S.; Husain, A.; Rehman, S.; Farooqui, A.;
Yusuf, M.A. Glycation and antioxidants: Hand in the glove of anti-
glycation and natural antioxidants. Curr. Protein Pept. Sci., 2020,
21(9), 899-915.
http://dx.doi.org/10.2174/1389203721666200210103304 PMID:
32039678
[18] Najafian, B.; Alpers, C.E.; Fogo, A.B. Pathology of human diabetic
nephropathy. Contrib. Nephrol., 2011, 170, 36-47.
http://dx.doi.org/10.1159/000324942 PMID: 21659756
[19] Bansode, S.; Bashtanova, U.; Li, R.; Clark, J.; Müller, K.H.;
Puszkarska, A.; Goldberga, I.; Chetwood, H.H.; Reid, D.G.; Col-
well, L.J.; Skepper, J.N.; Shanahan, C.M.; Schitter, G.; Mesquida,
P.; Duer, M.J. Glycation changes molecular organization and
charge distribution in type I collagen fibrils. Sci. Rep., 2020, 10(1),
3397.
http://dx.doi.org/10.1038/s41598-020-60250-9 PMID: 32099005
[20] Khanam, A.; Alouffi, S.; Rehman, S.; Ansari, I.A.; Shahab, U.;
Ahmad, S. An in vitro approach to unveil the structural alterations
in D -ribose induced glycated fibrinogen. J. Biomol. Struct. Dyn.,
2021, 39(14), 5209-5223.
http://dx.doi.org/10.1080/07391102.2020.1802339 PMID:
32772827
[21] Hudson, D.M.; Archer, M.; King, K.B.; Eyre, D.R. Glycation of
type I collagen selectively targets the same helical domain lysine
sites as lysyl oxidase–mediated cross-linking. J. Biol. Chem., 2018,
293(40), 15620-15627.
http://dx.doi.org/10.1074/jbc.RA118.004829 PMID: 30143533
[22] Xu, J.; Shi, G.P. Vascular wall extracellular matrix proteins and
vascular diseases. Biochim. Biophys. Acta Mol. Basis Dis., 2014,
1842(11), 2106-2119.
http://dx.doi.org/10.1016/j.bbadis.2014.07.008 PMID: 25045854
[23] Garcia-Fernandez, N.; Jacobs-Cachá, C.; Mora-Gutiérrez, J.M.;
Vergara, A.; Orbe, J.; Soler, M.J. Matrix metalloproteinases in dia-
betic kidney disease. J. Clin. Med., 2020, 9(2), 472.
http://dx.doi.org/10.3390/jcm9020472 PMID: 32046355
[24] Marshall, C.B. Rethinking glomerular basement membrane thick-
ening in diabetic nephropathy: Adaptive or pathogenic? Am. J.
Physiol. Renal Physiol., 2016, 311(5), F831-F843.
http://dx.doi.org/10.1152/ajprenal.00313.2016 PMID: 27582102
[25] Coughlan, M.T.; Forbes, J.M.; Cooper, M.E. Role of the AGE
crosslink breaker, alagebrium, as a renoprotective agent in diabetes.
Kidney Int., 2007, 72(106), S54-S60.
http://dx.doi.org/10.1038/sj.ki.5002387 PMID: 17653212
[26] Yin, Q.; Liu, H. Connective tissue growth factor and renal fibrosis.
Renal Fibrosis: Mech. Therap., 2019, 365-380.
[27] Husain, A.; Alouffi, S.; Khanam, A.; Akasha, R.; Khan, S.; Khan,
M. Non-inhibitory effects of the potent antioxidant from sp. on the
glycation reaction. Rev. Rom. Med. Lab., 2022, 30(2), 199-213.
[28] Yan, S.F.; Ramasamy, R.; Schmidt, A.M. The RAGE Axis. Circ.
Res., 2010, 106(5), 842-853.
http://dx.doi.org/10.1161/CIRCRESAHA.109.212217 PMID:
20299674
[29] Sun, L.; Kanwar, Y.S. Relevance of TNF-α in the context of other
inflammatory cytokines in the progression of diabetic nephropathy.
Kidney Int., 2015, 88(4), 662-665.
http://dx.doi.org/10.1038/ki.2015.250 PMID: 26422621
[30] Cannizzaro, L.; Rossoni, G.; Savi, F.; Altomare, A.; Marinello, C.;
Saethang, T.; Carini, M.; Payne, D.M.; Pisitkun, T.; Aldini, G.;
Leelahavanichkul, A. Regulatory landscape of AGE-RAGE-
oxidative stress axis and its modulation by PPARγ activation in
high fructose diet-induced metabolic syndrome. Nutr. Metab.
(Lond.), 2017, 14(1), 5.
http://dx.doi.org/10.1186/s12986-016-0149-z PMID: 28101123
[31] Tobon-Velasco, C Receptor for AGEs (RAGE) as mediator of NF-
kB pathway activation in neuroinflammation and oxidative stress.
CNS Neurol. Disord.-Drug Targets, 2014, 13(9), 1615-1626.
[32] Ramasamy, R.; Shekhtman, A.; Schmidt, A.M. The RAGE/DI-
APH1 signaling axis & implications for the pathogenesis of diabet-
ic complications. Int. J. Mol. Sci., 2022, 23(9), 4579.
http://dx.doi.org/10.3390/ijms23094579 PMID: 35562970
[33] Sanajou, D.; Ghorbani Haghjo, A.; Argani, H.; Aslani, S. AGE-
RAGE axis blockade in diabetic nephropathy: Current status and
future directions. Eur. J. Pharmacol., 2018, 833, 158-164.
http://dx.doi.org/10.1016/j.ejphar.2018.06.001 PMID: 29883668
[34] Alouffi, S.; Khanam, A.; Husain, A.; Akasha, R.; Rabbani, G.;
Ahmad, S. d-ribose-mediated glycation of fibrinogen: Role in the
induction of adaptive immune response. Chem. Biol. Interact.,
2022, 367, 110147.
http://dx.doi.org/10.1016/j.cbi.2022.110147 PMID: 36108717

Supplementary resource (1)

... Type 2 diabetes mellitus (T2D) is a critical global public health challenge as it raises the risk of all-cause mortality up to 15%, primarily due to cardiovascular complications [1]. Although the pathophysiology of T2D is complex, the non-enzymatic glycation reaction is one of the main factors in the progression and complications induced by diabetes [2]. Insulin resistance (IR), the origin of T2D, is intricately linked with atherogenic dyslipidemia, affecting triglyceride metabolism and both low-density lipoprotein (LDL) and high-density lipoprotein cholesterol (HDL-C) [3,4]. ...
Article
Full-text available
Evolocumab and empagliflozin yield a modest rise in plasma high-density lipopro-tein cholesterol (HDL-C) through unknown mechanisms. This study aims to assess the effect of evolocumab plus empagliflozin vs. empagliflozin alone on HDL subspecies isolated from individuals with type 2 diabetes mellitus (T2D). This post hoc prespecified analysis of the EXCEED-BHS3 trial compared the effects of a 16-week therapy with empagliflozin (E) alone or in combination with evolocumab (EE) on the lipid profile and cholesterol content in HDL subspecies in individuals with T2D divided equally into two groups of 55 patients. Both treatments modestly increased HDL-C. The cholesterol content in HDL subspecies 2a (7.3%), 3a (7.2%) and 3c (15%) increased from baseline in the E group, while the EE group presented an increase from baseline in 3a (9.3%), 3b (16%) and 3c (25%). The increase in HDL 3b and 3c was higher in the EE group when compared to the E group (p < 0.05). No significant interactive association was observed between changes in hematocrit and HDL-C levels after treatment. Over a 16-week period, empagliflozin with or without the addition of evolocumab led to a modest but significant increase in HDL-C. The rise in smaller-sized HDL particles was heterogeneous amongst the treatment combinations.
... Furthermore, numerous studies have shown the active involvement of ROS and AGEs in diabetes nephropathy, suggesting that they may serve as an early trigger for the morphological alterations seen in diabetes nephropathy [5,64]. In diabetes kidneys, ROS production has been shown to promote the synthesis of AGEs such as pentosidine and CML [65]. Previous studies have shown autoantibodies against D-ribose glycated hemoglobin (Hb), MG-glycated LDL, and D-ribose-induced DNA glycation in type 2 diabetes and its secondary complications [53,66,67]. ...
Article
Full-text available
The process of glycation, characterized by the non-enzymatic reaction between sugars and free amino groups on biomolecules, is a key contributor to the development and progression of both microvascular and macrovascular complications associated with diabetes, particularly due to persistent hyperglycemia. This glycation process gives rise to advanced glycation end products (AGEs), which play a central role in the pathophysiology of diabetes complications, including nephropathy. The d-ribose-mediated glycation of fibrinogen plays a central role in the pathogenesis of diabetes nephropathy (DN) and retinopathy (DR) by the generation and accumulation of advanced glycation end products (AGEs). Glycated fibrinogen with d-ribose (Rb-gly-Fb) induces structural changes that trigger an autoimmune response by generating and exposing neoepitopes on fibrinogen molecules. The present research is designed to investigate the prevalence of autoantibodies against Rb-gly-Fb in individuals with type 2 diabetes mellitus (T2DM), DN & DR. Direct binding ELISA was used to test the binding affinity of autoantibodies from patients' sera against Rb-gly-Fb and competitive ELISA was used to confirm the direct binding findings by checking the bindings of isolated IgG against Rb-gly-Fb and its native conformer. In comparison to healthy subjects, 32% of T2DM, 67% of DN and 57.85% of DR patients' samples demonstrated a strong binding affinity towards Rb-gly-Fb. Both native and Rb-gly-Fb binding by healthy subjects (HS) sera were non-significant (p > 0.05). Furthermore, the early, intermediate, and end products of glycation have been assessed through biochemical and physicochemical analysis. The biochemical markers in the patient groups were also significant (p < 0.05) in comparison to the HS group. This study not only establishes the prevalence of autoantibodies against d-ribose glycated fibrinogen in DN but also highlights the potential of glycated fibrinogen as a biomarker for the detection of DN and/or DR. These insights may open new avenues for research into novel therapeutic strategies and the prevention of diabetes-related nephropathy and retinopathy.
... 13 Alpha-amylase is an enzyme responsible for the breakdown of complex carbohydrates into simpler sugars, 14 whereas beta-glucosidase plays a crucial role in the final step of glucose release. 15,16 Inhibiting these enzymes can effectively regulate postprandial glucose levels, a critical factor in diabetes management. Cyanobacterial compounds have shown the ability to interact with these enzymes, suggesting their potential as inhibitors and regulators of carbohydrate digestion and glucose release. ...
Article
Full-text available
This research investigates the potential of bioactive compounds derived from cyanobacteria as inhibitors of alpha-amylase and beta-glucosidase enzymes, which are involved in starch digestion and glucose release. The study reveals strong molecular interactions between these compounds and the enzyme active sites through docking analysis. Notably, compounds such as Abietic, Anilide, Nostocarboline, Noscomin, Tanikolide, Tubercidin, Cryptophycin, and Cyanobacteria exhibit the lowest binding energies when interacting with alpha-amylase. Among them, Noscomin demonstrates the lowest docking score and binding energy against alpha-amylase, outperforming the reference compound metformin. Similarly, these compounds also display low binding energies when interacting with beta-glucosidase. The bioactive compounds from cyanobacteria show significant potential as inhibitors of alpha-amylase and beta-glucosidase, suggesting their efficacy in managing diabetes by slowing down starch digestion and controlling glucose release. Their superior binding affinities and lower binding energies, particularly Noscomin, indicate their potential in regulating blood sugar levels by interacting effectively with these enzymes. Thus, these compounds hold promise as valuable leads for developing alpha-amylase and beta-glucosidase inhibitors, contributing to the management of diabetes. Further research is required to understand the underlying mechanisms of action and assess the bioavailability, toxicity, and pharmacological potential of these cyanobacterial compounds. This investigation provides valuable insights into the potential of cyanobacterial bioactive compounds as effective candidates for the development of novel therapeutics targeting alpha-amylase and beta-glucosidase enzymes in diabetes management.
Article
Full-text available
Post-translational modifications (PTMs) are crucial epigenetic mechanisms that regulate various cellular biological processes. The use of mass spectrometry (MS)-proteomics has led to the discovery of numerous novel types of protein PTMs, such as acetylation, crotonylation, 2-hydroxyisobutyrylation, β-hydroxybutyrylation, protein propionylation and butyrylation, succinylation, malonylation, lactylation, and histone methylation. In this review, we specifically highlight the molecular mechanisms and roles of various histone and some non-histone PTMs in renal diseases, including diabetic kidney disease. PTMs exhibit diverse effects on renal diseases, which can be either protective or detrimental, depending on the specific type of protein PTMs and their respective targets. Different PTMs activate various signaling pathways in diverse renal pathological conditions, which could provide novel insights for studying epigenetic mechanisms and developing potential therapeutic strategies for renal diseases.
Article
Full-text available
When glucose and Amadori products are auto-oxidized, glycation occurs, resulting in the formation of early (Amadori) and late advanced glycation end products (AGEs), as well as free radicals. Glycation and an increase in free radical activity induce diabetic complications. Antioxidant and antiglycation compounds may aid in the prevention of oxidation and glycation. The goal of this study was to assess the antiglycation and antioxidant capacity of C-phycocyanin (C-PC) derived from Plectonema sp. The DPPH (1, 1-diphenyl-2-picrylhydrazyl), nitric oxide, hydroxyl radical scavenging activities and ferric ions reducing antioxidant power (FRAP) assays were used to assess antioxidant activity, while an in vitro bovine serum albumin-methyl glyoxal glycation (BSA-MG) model was used to assess glycation inhibitory potential. Glycation inhibition was measured using a variety of spectroscopic and biochemical parameters, including UV-visible & fluorescence spectroscopy, ketoamine, carbonyl and hydroxymethyl furfural content, as well as free lysine & free arginine estimations. In vitro, C-PC exhibited dose-dependent potent antioxidant activity, but lacked significant antiglycation potential. As a result, it is recommended that further studies be conducted to evaluate the antiglycation potential of C-PC.
Article
Full-text available
Increasing evidence links the RAGE (receptor for advanced glycation end products)/DIAPH1 (Diaphanous 1) signaling axis to the pathogenesis of diabetic complications. RAGE is a multi-ligand receptor and through these ligand–receptor interactions, extensive maladaptive effects are exerted on cell types and tissues targeted for dysfunction in hyperglycemia observed in both type 1 and type 2 diabetes. Recent evidence indicates that RAGE ligands, acting as damage-associated molecular patterns molecules, or DAMPs, through RAGE may impact interferon signaling pathways, specifically through upregulation of IRF7 (interferon regulatory factor 7), thereby heralding and evoking pro-inflammatory effects on vulnerable tissues. Although successful targeting of RAGE in the clinical milieu has, to date, not been met with success, recent approaches to target RAGE intracellular signaling may hold promise to fill this critical gap. This review focuses on recent examples of highlights and updates to the pathobiology of RAGE and DIAPH1 in diabetic complications.
Article
Full-text available
As of late, evidence has been emerging that the Maillard reaction (MR, also referred to as glycation) affects the structure and function of food proteins. MR induces the conformational and chemical modification of food proteins, not only on the level of IgG/IgE recognition, but also by increasing the interaction and recognition of these modified proteins by antigen-presenting cells (APCs). This affects their biological properties, including digestibility, bioavailability, immunogenicity, and ultimately their allergenicity. APCs possess various receptors that recognize glycation structures, which include receptor for advanced glycation end products (RAGE), scavenger receptors (SRs), galectin-3 and CD36. Through these receptors, glycation structures may influence the recognition, uptake and antigen-processing of food allergens by dendritic cells (DCs) and monocytes. This may lead to enhanced cytokine production and maturation of DCs, and may also induce adaptive immune responses to the antigens/allergens as a result of antigen uptake, processing and presentation to T cells. Here, we aim to review the current literature on the immunogenicity of AGEs originating from food (exogenous or dietary AGEs) in relation to AGEs that are formed within the body (endogenous AGEs), their interactions with receptors present on immune cells, and their effects on the activation of the innate as well as the adaptive immune system. Finally, we review the clinical relevance of AGEs in food allergies.
Article
Full-text available
C-phycocyanin (C-PC), the integral blue-green algae (BGA) constituent has been substantially delineated for its biological attributes. Numerous reports have illustrated differential extraction and purification techniques for C-PC, however, there exists paucity in a broadly accepted process of its isolation. In the present study, we reported a highly selective C-PC purification and characterization method from nontoxic, filamentous and non-heterocystous cyanobacterium Plectonema sp. C-PC was extracted by freeze-thawing, desalted and purified using ion-exchange chromatography. The purity of C-PC along with its concentration was found to be 4.12 and 245 µg/ml respectively. Comparative characterization of standard and purified C-PC was performed using diverse spectroscopic techniques namely Ultra Violet-visible, fluorescence spectroscopy and Fourier transform infrared (FT-IR). Sharp peaks at 620 nm and 350 nm with UV-visible and FT-IR spectroscopy respectively, confirmed amide I bands at around 1638 cm-1 (C=O stretching) whereas circular dichroism (CD) spectra exhibited α-helix content of secondary structure of standard 80.59% and 84.59% of column purified C-PC. SDS-PAGE exhibited two bands of α and β subunits 17 and 19 kDa respectively. HPLC evaluation of purified C-PC also indicated a close resemblance of retention peak time (1.465 min, 1.234 min, 1.097 min and 0.905 min) with standard C-PC having retention peak timing of 1.448 min, 1.233 min and 0.925 min. As a cautious approach, the purified C-PC was further lyophilized to extend its shelf life as compared to its liquid isoform. To evaluate the bioactive potential of the purified C-PC in silico approach was attempted. The molecular docking technique was carried out of C-PC as a ligand-protein with free radicals and α-amylase, α-glucosidase, glycogen synthase kinase-3 and glycogen phosphorylase enzymes as receptors to predict the free radical scavenging (antioxidant) and to target antidiabetic property of C-PC. In both receptors free radicals and enzymes, ligand C-PC plays an important role in establishing interactions within the cavity of active sites. These results established the antioxidant potential of C-PC and also give a clue towards its antidiabetic potential warranting further research.
Article
Full-text available
Plasma proteins persistently bear non-enzymatic post-translational modifications (NEPTM) that proceeds with nucleophilic addition between free amino groups of proteins, and carbonyl group of reducing sugars. Glycation, a prevalent NEPTM rush by the high availability of reducing sugars results in the generation of advanced glycation end products (AGEs). Plasma proteins are more vulnerable to glycation because of the presence of multiple glycation sites and are widely studied. However, fibrinogen glycation is less studied. Therefore, it was designed as an in vitro study to elucidate d-ribose mediated glycative damage suffered by fibrinogen protein at secondary and tertiary structure level. The glycation induced structural alterations were analyzed by UV-vis, fluorescence, circular dichroism, scanning electron microcopy and Fourier transform infrared spectroscopy. Glycation induced protein aggregation and fibrils formation was confirmed by thioflavin T and congo red assay. Moreover, molecular docking study was performed to further validate physicochemical characterization. Structural alterations, increased ketoamines, protein carbonyls and HMF contents were reported in d-ribose glycated fibrinogen against their native analogues. The results validate structural perturbations, increased glycoxidative stress and AGEs formation, which might influence normal function of fibrinogen especially blood coagulation cascade. Thus, we can conclude that under diabetes induced hyperglycemic state in physiological systems, d-ribose induced fibrinogen glycation might play a crucial role in the onset of micro- and macro-vascular complications, thereby worsen the diabetes associated secondary disorders. Moreover, this in vitro study might pave a path to choose fibrinogen as a future biomarker for the early detection of diabetes mediated vascular complications. Communicated by Ramaswamy H. Sarma
Article
Full-text available
Collagen fibrils are central to the molecular organization of the extracellular matrix (ECM) and to defining the cellular microenvironment. Glycation of collagen fibrils is known to impact on cell adhesion and migration in the context of cancer and in model studies, glycation of collagen molecules has been shown to affect the binding of other ECM components to collagen. Here we use TEM to show that ribose-5-phosphate (R5P) glycation of collagen fibrils – potentially important in the microenvironment of actively dividing cells, such as cancer cells – disrupts the longitudinal ordering of the molecules in collagen fibrils and, using KFM and FLiM, that R5P-glycated collagen fibrils have a more negative surface charge than unglycated fibrils. Altered molecular arrangement can be expected to impact on the accessibility of cell adhesion sites and altered fibril surface charge on the integrity of the extracellular matrix structure surrounding glycated collagen fibrils. Both effects are highly relevant for cell adhesion and migration within the tumour microenvironment.
Article
Full-text available
Background: Health system planning requires careful assessment of chronic kidney disease (CKD) epidemiology, but data for morbidity and mortality of this disease are scarce or non-existent in many countries. We estimated the global, regional, and national burden of CKD, as well as the burden of cardiovascular disease and gout attributable to impaired kidney function, for the Global Burden of Diseases, Injuries, and Risk Factors Study 2017. We use the term CKD to refer to the morbidity and mortality that can be directly attributed to all stages of CKD, and we use the term impaired kidney function to refer to the additional risk of CKD from cardiovascular disease and gout. Methods: The main data sources we used were published literature, vital registration systems, end-stage kidney disease registries, and household surveys. Estimates of CKD burden were produced using a Cause of Death Ensemble model and a Bayesian meta-regression analytical tool, and included incidence, prevalence, years lived with disability, mortality, years of life lost, and disability-adjusted life-years (DALYs). A comparative risk assessment approach was used to estimate the proportion of cardiovascular diseases and gout burden attributable to impaired kidney function. Findings: Globally, in 2017, 1·2 million (95% uncertainty interval [UI] 1·2 to 1·3) people died from CKD. The global all-age mortality rate from CKD increased 41·5% (95% UI 35·2 to 46·5) between 1990 and 2017, although there was no significant change in the age-standardised mortality rate (2·8%, -1·5 to 6·3). In 2017, 697·5 million (95% UI 649·2 to 752·0) cases of all-stage CKD were recorded, for a global prevalence of 9·1% (8·5 to 9·8). The global all-age prevalence of CKD increased 29·3% (95% UI 26·4 to 32·6) since 1990, whereas the age-standardised prevalence remained stable (1·2%, -1·1 to 3·5). CKD resulted in 35·8 million (95% UI 33·7 to 38·0) DALYs in 2017, with diabetic nephropathy accounting for almost a third of DALYs. Most of the burden of CKD was concentrated in the three lowest quintiles of Socio-demographic Index (SDI). In several regions, particularly Oceania, sub-Saharan Africa, and Latin America, the burden of CKD was much higher than expected for the level of development, whereas the disease burden in western, eastern, and central sub-Saharan Africa, east Asia, south Asia, central and eastern Europe, Australasia, and western Europe was lower than expected. 1·4 million (95% UI 1·2 to 1·6) cardiovascular disease-related deaths and 25·3 million (22·2 to 28·9) cardiovascular disease DALYs were attributable to impaired kidney function. Interpretation: Kidney disease has a major effect on global health, both as a direct cause of global morbidity and mortality and as an important risk factor for cardiovascular disease. CKD is largely preventable and treatable and deserves greater attention in global health policy decision making, particularly in locations with low and middle SDI. Funding: Bill & Melinda Gates Foundation.
Article
Full-text available
The non-enzymatic interaction of sugar and protein resulting in the formation of advanced glycation end products responsible for cell signaling alterations ultimately lead to the human chronic disorders such as diabetes mellitus, cardiovascular diseases, cancer, etc. Studies suggest that AGEs upon interaction with receptors for advanced glycation end products (RAGE) result in the production of pro-inflammatory molecules and free radicals that exert altered gene expression effect. Till date many studies unveiled the potent role of synthetic and natural agents in inhibiting the glycation reaction at lesser or greater extent. This review focuses on the hazards of glycation reaction and its inhibition by natural antioxidants including polyphenols.
Article
A nonenzymatic reaction between reducing sugars and amino groups of proteins results in the formation of advanced glycation end products, which are linked to a number of chronic progressive diseases with macro- and microvascular complications. In this research, we sought to ascertain the immunological response to d-ibose-glycated fibrinogen. New Zealand White female rabbits were immunized with native and d-ribose-glycated (Rb-gly-Fb) fibrinogen and used for studying the immunological response. Serum from these rabbits analyzed using direct binding and competitive inhibition ELISA was found to contain a high titer of antibodies against Rb-gly-Fb; Rb-gly-Fb was much more immunogenic than its native form. The IgG against Rb-gly-Fb (Rb-gly-Fb-IgG) was highly specific against the immunogenic protein. Moreover, histopathology and immunofluorescence studies revealed the deposition of the Rb-gly-Fb-IgG immune complex in the glomerular basement membrane of the kidneys of immunized rabbits. Furthermore, immunization with Rb-gly-Fb increased the expression of genes encoding proinflammatory cytokines, tumour necrosis factor α, interleukin-6, interleukin-1β, and interferon-gamma, which is indicative of increased inflammation and the antigenic role of Rb-gly-Fb in provoking an immune response.
Article
One of the hallmark features in the neurodegenerative disorders (NDDs) is the accumulation of aggregated and/or non-functional protein in the cellular milieu. Post-translational modifications (PTMs) are an essential regulator of non-functional protein aggregation in the pathogenesis of NDDs. Any alteration in the post-translational mechanism and the protein quality control system, for instance, molecular chaperone, ubiquitin-proteasome system, autophagy-lysosomal degradation pathway, enhances the accumulation of misfolded protein, which causes neuronal dysfunction. Post-translational modification plays many roles in protein turnover rate, accumulation of aggregate and can also help in the degradation of disease-causing toxic metabolites. PTMs such as acetylation, glycosylation, phosphorylation, ubiquitination, palmitoylation, SUMOylation, nitration, oxidation, and many others regulate protein homeostasis, which includes protein structure, functions and aggregation propensity. Different studies demonstrated the involvement of PTMs in the regulation of signaling cascades such as PI3K/Akt/GSK3β, MAPK cascade, AMPK pathway, and Wnt signaling pathway in the pathogenesis of NDDs. Further, mounting evidence suggests that targeting different PTMs with small chemical molecules, which acts as an inhibitor or activator, reverse misfolded protein accumulation and thus enhances the neuroprotection. Herein, we briefly discuss the protein aggregation and various domain structures of different proteins involved in the NDDs, indicating critical amino acid residues where PTMs occur. We also describe the implementation and involvement of various PTMs on signaling cascade and cellular processes in NDDs. Lastly, we implement our current understanding of the therapeutic importance of PTMs in neurodegeneration, along with emerging techniques targeting various PTMs.