Background: Kidney transplantation was an effective method for treating chronic kidney failure via transplanting a healthy kidney from a donor to a patient with the loss of kidney function. However, clinical studies revealed that the posttransplantation status of patients was associated with a substantial aggregation of risk factors contributing to metabolic syndrome. Summary: This article provided a comprehensive review of the current researches on metabolic syndrome after kidney transplantation, and the latest advances in the interaction between metabolism and immune cells were also covered. Key Messages: Our aim was to identify and intervene high-risk recipients in time and thus improving the prognosis of recipients.

Chronic kidney disease (CKD) is characterized by progressive structural and functional impairment of the kidneys due to various etiologies, and chronic kidney failure represented the ultimate outcome of kidney-related conditions [1, 2]. CKD has gradually become a worldwide health burden. In 2020, the Lancet published a report on the global epidemiology of CKD, revealing a global CKD patient population of 697.5 million (including 132 million in China). CKD led to 1.2 million deaths worldwide and was expected to increase to 2.2 million deaths by 2040, and this number may rise to 4 million in severe cases [3]. Chronic kidney failure often remained undetected until it reached an advanced stage due to its insidious onset, posing a grave threat to the patient’s well-being, and ultimately, the vast majority of individuals afflicted by this ailment necessitated life-sustaining measures such as dialysis or kidney transplantation. For end-stage kidney failure patients, kidney transplantation was the most optimal approach for enhancing life quality [4]. However, clinical investigation revealed that the long-term life quality of kidney transplantation recipients was unsatisfactory.

The state following kidney transplantation aggregated numerous risk factors for various diseases such as metabolic disturbances in glucose and lipid metabolism, hyperuricemia, and hypertension, which collectively contributed to metabolic syndrome, a clinical syndrome with various metabolic disorders such as obesity, glucose metabolism disturbances, lipid metabolic irregularities, and hypertension. It was discerned that the incidence of various diseases was higher in kidney transplantation recipients than the general population [5]. In comparison to the non-metabolic syndrome group, the occurrence of metabolic syndrome was correlated with an increased risk of graft loss. The jeopardy of graft loss escalated in tandem with the augmented quantity of dysfunctional metabolic syndrome components [6]. Although there might be debates surrounding the definition and diagnosis of metabolic syndrome, it was generally accepted that the components associated with metabolic syndrome served as significant risk factors for cardiovascular and cerebrovascular events [7]. Thus, analyzing the risk factors, early identifying, and intervening with high-risk recipients were of great significance in reducing the incidence of metabolic syndrome after kidney transplantation. This article provided a comprehensive review of the current researches on post-kidney transplantation metabolic syndrome, thus offering a better understanding of metabolic syndrome and enhancing life quality of the recipients.

Metabolic syndrome was not a distinct ailment but rather a constellation of various metabolic risk factors. Although the precise definition of metabolic syndrome remained a subject of debate, it was widely acknowledged that nearly all metabolic aberrations, including but not limited to obesity, lipid disturbances, hypertension, and insulin resistance, were regarded as potential unified pathogenic elements. The incidence of metabolic syndrome was typically threefold than that of diabetes. According to the IDF Diabetes Atlas, as of 2015, the global prevalence of diabetes was 8.8% (415 million), with projections indicating an increase to 10.4% (642 million) by 2040. Consequently, it was estimated that the global prevalence of metabolic syndrome affected approximately one-fourth of the world’s population, suggesting that over one billion people worldwide were presently subject to the influence of metabolic syndrome. Some epidemiological studies have identified that lifestyle habits and socio-economic status significantly influenced the prevalence of metabolic syndrome in gender, age, and racial/ethnic groups [8, 9]. The precise etiology of metabolic syndrome still remained not entirely comprehended [10, 11]. Abdominal obesity and insulin resistance were commonly considered pivotal factors contributing to the development of metabolic syndrome [12]. Genetic susceptibility, pro-inflammatory status, leptin, physical inactivity, adiponectin, thyroid hormones, aging, and more could also serve as pathological elements. Recently, a research reported that macrophages within adipose tissues played a central role in energy metabolism, M1 macrophages promoted hepatic fibrosis and fat synthesis, while anti-inflammatory M2 macrophages exhibited a macrophage restriction defect in a mouse model and resulted in hepatic fibrosis and obesity phenotypes [13]. Endoplasmic reticulum stress represented another pivotal factor in the pathogenesis of metabolic syndrome. In murine experiments, Shan and colleagues demonstrated that endoplasmic reticulum stress induced by a high-fat diet was dependent on the activity of inositol-requiring enzyme 1 alpha (IRE1alpha), an enzyme crucial for sulfur-free protein synthesis. Lack of IRE1alpha activity prevented obesity, insulin resistance, and hepatic fibrosis induced by high-fat diet. IRE1alpha also inhibited M2 macrophages within adipose tissues [14]. Epigenetics appeared to play a more substantial role in promoting metabolic syndrome. Offspring born to parents afflicted by obesity tended to inherit the same condition [15]. As the cellular and molecular mechanisms underlying the pathogenesis of metabolic syndrome remained not entirely elucidated, treatment regimens were yet to be established. Currently available treatment strategies primarily included promoting a physically active and healthful lifestyle or focusing on addressing the individual components of metabolic syndrome [16]. The overarching objective was to mitigate the risks of cardiovascular disease and type 2 diabetes or prevent their onset [17].

Current State of Research on Hypertension Post-Kidney Transplantation

The most prevalent postoperative complication in organ transplantation was hypertension. Aberrations in postoperative blood pressure were significantly related to the incidence of cardiovascular events. If left unregulated, they substantially elevated the risk of cardiovascular complications. Scholars have believed that arterial hypertension following kidney transplantation was intricately linked to suboptimal cardiovascular and renal outcomes [18], which was accompanied by kidney function impairment, diminished graft survival rates, and heightened mortality. The proportion of postoperative kidney transplantation recipients with systolic blood pressure exceeding 140 mm Hg (10 mm Hg = 1.33 kPa) ranged from 55.5% to 90.0% [19], leading to approximately 60–70% of transplanted kidneys experiencing varying degrees of impairment. Various factors contributed to hypertension in organ transplantation recipients. Protein encoded by CYP3A5 gene played a significant role in sodium and aldosterone metabolism in kidney, which also amplified the hypertensive effects of calcium channel blockers [20]. Podocalyxin-1 was a pivotal intracellular protein governing the endocytic machinery [21] and played a leading role in regulating the degradation of transforming growth factor-β. In the absence of podocalyxin-1, there was a significant increase in transforming growth factor-β activity in donors, accelerating the abnormal interstitial fibrosis process in transplanted kidneys [21]. This cascade contributed to hypertension and the deterioration of kidney transplantation. Posttransplantation hypertension might be a principal etiology of metabolic syndrome, which, in turn, exacerbated hypertension, forming a deleterious feedback loop [22].

Current State of Research on Post-Kidney Transplantation Dyslipidemia

Dyslipidemia was the disorder of lipid metabolism in human body, which was characterized by the abnormal levels of total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C), and was regarded as an autonomous risk factor for cardiovascular events. In prospective kidney transplantation candidates, compared to transplantation recipients, there was a significant increase in the prevalence of hypercholesterolemia, increasing from 25% to 60% [23]. The overall elevation in total cholesterol was approximately 27%. The pathogenic mechanisms of posttransplantation dyslipidemia were multifaceted [24, 25]. Factors such as chronic anemia, elevated homocysteine levels, chronic inflammation, proteinuria, diuretics, and β-adrenergic blockers were recognized contributors to lipid abnormalities and cardiovascular diseases. Additionally, sedentariness, the excessive intake of saturated fats, cholesterol, and trans fats were closely associated with lipid disorder [26]. It has been reported that immunosuppressants played a pivotal role in lipid abnormality [27]. Cyclosporine, tacrolimus, sirolimus, everolimus, corticosteroids, as well as a limited quantity of thiopurines and mycophenolic acid were all associated with lipid imbalance. Cyclosporine decreased LDL receptors expression by increasing proprotein convertase subtilisin/kexin type 9 (PCSK9) and inhibited cholesterol transport to the intestines by reducing cholesterol expression [28]. Conducting mechanistic researches to identify effective targets and corresponding medications for kidney transplantation recipients was significant.

Current State of Research on Post-Kidney Transplantation Hyperuricemia

Hyperuricemia was a metabolic disorder characterized by excessive uric acid production or reduced excretion due to the abnormal purine metabolism, as well as the elevated blood uric acid levels resulted from the excessive intake of exogenous purines. Hyperuricemia not only impacted kidney transplantation function but also elevated the risk of cardiovascular diseases, which was a significant risk factor affecting the long-term survival of transplanted kidneys. Epidemiological investigations revealed a rising prevalence of hyperuricemia, ranging from 10% to 15% in the general population. In China, the incidence of hyperuricemia has reached 20% in coastal regions and developed cities, reaching the level of developed countries [29, 30]. The incidence of hyperuricemia in kidney transplantation recipients was significantly higher than that of the general population, affecting approximately 40–60% of recipients [31]. Moreover, a study involving 9,589 participants suggested that incorporating uric acid into the definition of metabolic syndrome was crucial when assessing the risk of mortality in kidney transplantation recipients [32]. Although the pathophysiological molecular mechanisms of kidney damage caused by hyperuricemia were not yet fully understood, current researches suggested the involvement of oxidative stress, endothelial dysfunction, renal fibrosis, and inflammation. Extracellular matrix played a pivotal role in the progression of interstitial fibrosis in the kidney, epithelial-mesenchymal transition (EMT) promoted Extracellular matrix production [33]. Researches revealed that the TGF-β/Smad3 pathway played a pivotal role in EMT. Hyperuricemia resulted in EMT and renal tubular injury in mice through the modulation of the Wnt5a/Ror2 or TLR4/NF-κB signaling pathways. EMT has been regarded as a novel therapeutic target owing to its reversibility [34, 35].

Current Status of Research on Post-Kidney Transplant Diabetes

Posttransplantation diabetes mellitus (PTDM) denoted the onset of diabetes following organ transplantation. The incidence of PTDM ranged from 2% to 50%. The substantial variation in incidence was attributed to the differing criteria adopted by various research centers and disparities in posttransplantation immunosuppressive regimens [36]. Risk factors for PTDM in kidney transplantation recipients were categorized into 2 classes: transplantation-related, encompassing the use of glucocorticoids, CNIs, viral infections, posttransplantation weight gain, and non-transplantation-related, including gender, age, ethnicity, obesity, family history of diabetes, and metabolic syndrome. The pathogenic mechanisms of PTDM were similar to type 2 diabetes mellitus [37], both exhibiting the decreased insulin sensitivity, augmented peripheral insulin resistance, weakened insulin secretion by pancreatic β cells, and reduced glucose tolerance. Some scholars believed that the evolution and pathogenic mechanisms of PTDM conformed to pre-transplantation β-cell impairment [38]. Pre-diabetic patients might progress to late-stage PTDM. Individuals with normal sugar metabolism might also experience the onset of new diabetes [39]. In summary, post-kidney transplantation patients experienced metabolic disruptions in fundamental substances, including sugars, lipids, proteins, purines, amino acids, electrolytes, posing a significant threat to life quality.

Warburg Effect and Activation of Immune Cells

Until now, most researches on immunometabolism focused on how immune cells simultaneously and differentially used nutrients to meet metabolic demands associated with effector function and differentiation states. In essence, environments influenced the metabolism and function of immune cells. The central metabolic program for the activation of innate and adaptive immune cells was glycolysis, often referred to as the Warburg effect [40]. During this activation process, innate immune cells and T cells, which originally mainly relied on oxidative phosphorylation for energy supply, rapidly switched to aerobic glycolysis (Warburg effect) for energy supply, thus quickly meeting the increased energy demands and providing key metabolic substrates. Hence, glucose exerted a significant effect on the metabolism of immune cells, and interfering with the Warburg effect of immune cells will affect their activation, differentiation, and memory formation. Metabolic processes, such as glycolysis and mitochondrial oxidative phosphorylation, governed the transformation of precursor cells into pathogenic and regulatory T (Treg) cells [41]. Helper T cells exhibited the enhanced glycolysis, relying on the glucose transporter protein glucose transporter1 (GLUT1), to facilitate the effector functions of Th1 and Th17 cells [42]. Modulation of GLUT1-mediated glycolysis in Treg cells, in turn, governed the equilibrium between proliferation and suppressive capabilities. It has also been reported that genes encoding amino acid transport proteins were the most upregulated genes after T-cell activation [43], suggesting that amino acid exchange solution was a critical aspect of immune regulation. In other words, apart from glucose, different amino acids also impacted the metabolism of immune cells.

Glutamine

Glutamine exerted multiple biological roles. In proliferating cells, glutamine provided nitrogen elements for amino acid and nucleic acid synthesis and offered carbon elements to replenish tricarboxylic acid (TCA) cycle intermediates, which was the cornerstone of biosynthesis. Experimental evidence indicated that restricting glutamine inhibited the effector function of differentiated CD8+ T cells [44]. Restricting glutamine during the activation of CD8+ T cells resulted in the differentiation into a long-term, persisting memory phenotype [45]. Metabolic byproducts of glutamine also modulated various cellular functions. Under the influence of MYC, glutamine was metabolized by glutaminase into glutamate, which was further converted into α-ketoglutarate by glutamate dehydrogenase before entering the TCA cycle [46]. α-Ketoglutarate and other metabolic products of the TCA cycle, such as succinate and fumarate, modulated the activity of numerous cellular factors, including epigenetic remodeling and the stability of crucial transcription factors like HIF-1α [47]. Furthermore, T cells and macrophages exhibited the capacity to catabolize glutamine to enhance the generation of ATP [48]. The inhibitory effect of glutaminase (referring to the conversion of glutamine to glutamate) impaired the proliferation and activation of CD4+ T cells and CD8+ T cells, with minimal interference in the generation of most cellular effector molecules. The same inhibitory action suppressed the differentiation of Th17 cells [49].

Methionine and Serine

Researches have shown that methionine played a crucial role in immune cells. Controlling cellular methionine levels modulated the state of T cells. When T cells were stimulated by antigens, they maintained the transport of methionine into the cells, thus increasing its intracellular concentration, which was the rate-limiting step where intracellular methionine supplied the methyl groups to the targets to be methylated [50]. Methionine provided methyl groups by generating S-adenosyl methionine (SAM). SAM served as the methyl donor for DNA, RNA, and protein methyltransferases [51]. Only T cells with an ample intracellular supply of methionine promoted DNA methylation, protein synthesis, and proliferation. Serine, a nonessential amino acid, enhanced T-cell proliferation and immune function, which was directly absorbed by cells or synthesized de novo from glycolytic metabolite 3-phosphoglycerate (3-PG) [52]. Serine also served as a methyl donor, providing hydroxymethyl as a one-carbon unit of tetrahydrofolate. Intracellular serine accumulation stimulated one-carbon metabolism, leading to the proliferation of crucial Treg cells that underlie self-immune tolerance. The extracellular availability of serine and methionine influenced the proliferation of T lymphocytes through actions related to one-carbon metabolism [53, 54]. Within T cells, the divergence of folate and methionine cycles coupled with the fact that carbon derived from serine did not integrate into the methionine cycle; external methionine may indeed stand as the exclusive origin of the methyl moiety in SAM. This phenomenon significantly affected subsequent methylation reactions. Serine also emerged as a pivotal switch governing the regulation of both T regulatory cells and effector T cells, a factor of substantial importance in the induction of posttransplantation immune tolerance.

Additional Amino Acids

It has been reported that supraphysiological levels of arginine suppressed T-cell-mediated cytokine production and enhanced T-cell survival [55]. The consumption of tryptophan impeded the functionality of T cells by activating integrated stress responses [56]. A recent study revealed that the availability of alanine impacted protein synthesis during the early stage of T-cell activation [57]. These findings suggested that various amino acids exerted distinct cell-specific effects on immune cells and provided the potential for targeting amino acid metabolism in different immune cells as a therapeutic approach in human diseases.

In conclusion, immunosuppressive drugs, obesity, diabetes mellitus, hypertension, albuminuria, improper diets, lack of exercises, and age were all risk factors for posttransplantation metabolic syndrome of recipients (Table 1); monitoring these components before and after the operation in kidney transplantation recipients may benefit clinical practices. However, this article exhibited certain limitations. Abdominal circumference data were not included. Although body mass index served as a critical value for diagnosing obesity, abdominal circumference data would enhance the reliability of the results. Additionally, sufficient Asian models were not collected for clinical external validation due to time constraints. In the future, large-scale studies or interventional studies will be conducted to provide a basis for reducing the occurrence of metabolic syndrome after kidney transplantation. Also, targeted corrections of the host’s metabolic environments hold the potential to enhance the induction of immune tolerance by regulating immune cells, and the clinical detection on the changes of amino acid levels in kidney transplantation recipients was necessary.

Table 1.

Risk factors of metabolic syndrome

SymptomAuthorYearIncidence rateRaceRisk factors
Metabolic syndrome Sharif et al. [582010 43% Britain Immunosuppressive drugs, obesity, diabetes mellitus, hypertension, albuminuria, improper diets, lack of exercises, unhealthy lifestyles, age, sex 
Metabolic syndrome Luan et al. [592010 53.1% America 
Metabolic syndrome Hami et al. [602017 52.8% Iran 
Metabolic syndrome Lee et al. [612023 40.1% Korea 
Hypertension Korogiannou et al. [622021 88.3% Greece Hypertension before transplantation, donor hypertension, preservation of the primary kidney, obesity, decreased renal flow, smoking, diabetes mellitus, immunosuppressive drugs 
Hypertension Arabi et al. [632023 79.2% Saudi Arabia 
Hypertension Speer et al. [642024 77% Multiple countries 
Dyslipidemia Numakura et al. [652015 34.9% Japan Immunosuppressive drugs, hypertension, diabetes mellitus, age, sex, genetic factors, history of cardiovascular diseases 
Dyslipidemia Borda et al. [662011 22% Hungary 
PTDM Joss et al. [672007 7.7% Britain Age, obesity, family history, high blood glucose before transplantation, history of tacrolimus use, hypertension, polycystic kidney, ethnic group, low physical activities, complications after renal transplantation 
PTDM Munshi et al. [682020 19% America 
PTDM Schweer et al. [692014 16.7% Germany 
PTDM Paek et al. [702019 11.8% Korea 
PTDM Du et al. [712024 20% Multiple countries 
Hyperuricemia Mazzali et al. [722014 44.13% Brazil Calcineurin inhibitors, obesity, sex, age, diuretics, eating habits, preoperative hyperuricemia 
SymptomAuthorYearIncidence rateRaceRisk factors
Metabolic syndrome Sharif et al. [582010 43% Britain Immunosuppressive drugs, obesity, diabetes mellitus, hypertension, albuminuria, improper diets, lack of exercises, unhealthy lifestyles, age, sex 
Metabolic syndrome Luan et al. [592010 53.1% America 
Metabolic syndrome Hami et al. [602017 52.8% Iran 
Metabolic syndrome Lee et al. [612023 40.1% Korea 
Hypertension Korogiannou et al. [622021 88.3% Greece Hypertension before transplantation, donor hypertension, preservation of the primary kidney, obesity, decreased renal flow, smoking, diabetes mellitus, immunosuppressive drugs 
Hypertension Arabi et al. [632023 79.2% Saudi Arabia 
Hypertension Speer et al. [642024 77% Multiple countries 
Dyslipidemia Numakura et al. [652015 34.9% Japan Immunosuppressive drugs, hypertension, diabetes mellitus, age, sex, genetic factors, history of cardiovascular diseases 
Dyslipidemia Borda et al. [662011 22% Hungary 
PTDM Joss et al. [672007 7.7% Britain Age, obesity, family history, high blood glucose before transplantation, history of tacrolimus use, hypertension, polycystic kidney, ethnic group, low physical activities, complications after renal transplantation 
PTDM Munshi et al. [682020 19% America 
PTDM Schweer et al. [692014 16.7% Germany 
PTDM Paek et al. [702019 11.8% Korea 
PTDM Du et al. [712024 20% Multiple countries 
Hyperuricemia Mazzali et al. [722014 44.13% Brazil Calcineurin inhibitors, obesity, sex, age, diuretics, eating habits, preoperative hyperuricemia 

All authors declare no conflict of interest.

This work was supported by the Natural Science Fund Program of Science and Technology Department of Jilin Province (No. 20220101329JC), Jilin Province College Students Innovation Training Project (No. s202313706036, 2023-JYSC039), Jilin Medical University College Students Innovation Training Project (No. 2024CXXL014), and Science and Technology Program of Education Department of Jilin Province (No. JJKH20230543KJ).

Kejing Zhu and Yuji Jin conceived the idea of this study. Kejing Zhu, Yuji Jin, Cheng Wen, Xinrui Zheng, Zhixiong Li, Yunjian Chen, Yulin Niu, and Wei Pan assisted in the literature search. Yong Jiang wrote the final report. Yingji Ji reviewed and revised the article. Weijian Liu contributed to the revision. All authors read and approved the final manuscript.

Additional Information

Kejing Zhu and Yuji Jin contributed equally to this work.

1.
Webster
AC
,
Nagler
EV
,
Morton
RL
,
Masson
P
.
Chronic kidney disease
.
Lancet
.
2017
;
389
(
10075
):
1238
52
.
2.
Lv
JC
,
Zhang
LX
.
Prevalence and disease burden of chronic kidney disease
.
Adv Exp Med Biol
.
2019
;
1165
:
3
15
.
3.
GBD Chronic Kidney Disease Collaboration
.
Global, regional, and national burden of chronic kidney disease, 1990-2017: a systematic analysis for the global burden of disease study 2017
.
Lancet
.
2020
;
395
(
10225
):
709
33
.
4.
De Brito
DCS
,
Machado
EL
,
Reis
IA
,
Moreira
DP
,
Nébias
THM
,
Cherchiglia
ML
.
Modality transition on renal replacement therapy and quality of life of patients: a 10-year follow-up cohort study
.
Qual Life Res
.
2019
;
28
(
6
):
1485
95
.
5.
Coniglio
AC
,
Bryner
BS
,
Devore
AD
,
Patel
CB
.
Trends in cardiovascular medicine: update on cardiac transplantation
.
Trends Cardiovasc Med
.
2023
;
33
(
1
):
46
50
.
6.
Lee
YH
,
Song
SH
,
Song
S
,
Shin
H
,
Yang
J
,
Kim
M
, et al
.
Clinical implications of changes in metabolic syndrome status after kidney transplantation: a nationwide prospective cohort study
.
Kidney Int Rep
.
2023
;
8
(
3
):
389
.
7.
Fändriks
L
.
Roles of the gut in the metabolic syndrome: an overview
.
J Intern Med
.
2017
;
281
(
4
):
319
36
.
8.
Ogurtsova
K
,
da Rocha Fernandes
JD
,
Huang
Y
,
Linnenkamp
U
,
Guariguata
L
,
Cho
NH
, et al
.
Idf diabetes atlas: global estimates for the prevalence of diabetes for 2015 and 2040
.
Diabetes Res Clin Pract
.
2017
;
128
:
40
50
.
9.
Elabbassi
WN
,
Haddad
HA
.
The epidemic of the metabolic syndrome
.
Saudi Med J
.
2005
;
26
(
3
):
373
5
.
10.
Aguilar
M
,
Bhuket
T
,
Torres
S
,
Liu
B
,
Wong
RJ
.
Prevalence of the metabolic syndrome in the United States, 2003-2012
.
JAMA
.
2015
;
313
(
19
):
1973
4
.
11.
Cornier
MA
,
Dabelea
D
,
Hernandez
TL
,
Lindstrom
RC
,
Steig
AJ
,
Stob
NR
, et al
.
The metabolic syndrome
.
Endocr Rev
.
2008
;
29
(
7
):
777
822
.
12.
Wang
HH
,
Lee
DK
,
Liu
M
,
Portincasa
P
,
Wang
QH
.
Novel insights into the pathogenesis and management of the metabolic syndrome
.
Pediatr Gastroenterol Hepatol Nutr
.
2020
;
23
(
3
):
189
230
.
13.
Jing
Y
,
Wu
F
,
Li
D
,
Yang
L
,
Li
Q
,
Li
R
.
Metformin improves obesity-associated inflammation by altering macrophages polarization
.
Mol Cell Endocrinol
.
2018
;
461
:
256
64
.
14.
Shan
B
,
Wang
X
,
Wu
Y
,
Xu
C
,
Xia
Z
,
Dai
J
, et al
.
The metabolic er stress sensor ire1α suppresses alternative activation of macrophages and impairs energy expenditure in obesity
.
Nat Immunol
.
2017
;
18
(
5
):
519
29
.
15.
Waalen
J
.
The genetics of human obesity
.
Transl Res
.
2014
;
164
(
4
):
293
301
.
16.
Mottillo
S
,
Filion
KB
,
Genest
J
,
Joseph
L
,
Pilote
L
,
Poirier
P
, et al
.
The metabolic syndrome and cardiovascular risk: a systematic review and meta-analysis
.
J Am Coll Cardiol
.
2010
;
56
(
14
):
1113
32
.
17.
Keaney
JF
,
Curfman
GD
,
Jarcho
JA
.
A pragmatic view of the new cholesterol treatment guidelines
.
N Engl J Med
.
2014
;
59
(
4
):
1174
5
.
18.
Weir
MR
,
Burgess
ED
,
Cooper
JE
,
Fenves
AZ
,
Goldsmith
D
,
Mckay
D
, et al
.
Assessment and management of hypertension in transplant patients
.
J Am Soc Nephrol
.
2015
;
26
(
6
):
1248
60
.
19.
Rebelo
RNS
,
Rodrigues
CIS
.
Arterial hypertension in kidney transplantation: huge importance, but few answers
.
J Bras Nefrol
.
2023
;
45
(
1
):
84
94
.
20.
Renders
L
,
Frisman
M
,
Ufer
M
,
Mosyagin
I
,
Haenisch
S
,
Ott
U
, et al
.
Cyp3a5 genotype markedly influences the pharmacokinetics of tacrolimus and sirolimus in kidney transplant recipients
.
Clin Pharmacol Ther
.
2007
;
81
(
2
):
228
34
.
21.
Hesselink
D
,
Bouamar
R
,
Elens
L
,
van Schaik
RHN
,
van Gelder
T
.
The role of pharmacogenetics in the disposition of and response to tacrolimus in solid organ transplantation
.
Clin Pharmacokinet
.
2014
;
53
(
2
):
123
39
.
22.
Palanisamy
A
,
Reeves-Daniel
AM
,
Freedman
BI
.
The impact of apol1 , cav1, and abcb1 gene variants on outcomes in kidney transplantation: donor and recipient effects
.
Pediatr Nephrol
.
2014
;
29
(
9
):
1485
92
.
23.
Joint Committee for Developing Chinese guidelines on Prevention and Treatment of Dyslipidemia in Adults
.
Chinese guidelines on prevention and treatment of dyslipidemia in adults
.
Zhonghua Xin Xue Guan Bing Za Zhi
.
2007
;
35
(
5
):
390
419
.
24.
Ojo
AO
.
Cardiovascular complications after renal transplantation and their prevention
.
Transplantation
.
2006
;
82
(
5
):
603
11
.
25.
Corsini
A
.
The safety of HMG-CoA reductase inhibitors in special populations at high cardiovascular risk
.
Cardiovasc Drugs Ther
.
2003
;
17
(
3
):
265
85
.
26.
Karabulut
U
,
Çakır
Ü
.
Non-HDL cholesterol is an independent predictor of long-term cardiovascular events in patients with dyslipidemia after renal transplantation
.
Int J Clin Pract
.
2021
;
75
(
9
):
e14465
.
27.
Arnett
DK
,
Blumenthal
RS
,
Albert
MA
,
Buroker
AB
,
Goldberger
ZD
,
Hahn
EJ
, et al
.
2019 ACC/AHA guideline on the primary prevention of cardiovascular disease: executive summary: a report of the american college of cardiology/american heart association task force on clinical practice guidelines
.
Circulation
.
2019
;
140
:
e563
-
95
.
28.
Ponticelli
C
,
Arnaboldi
L
,
Moroni
G
,
Corsini
A
.
Treatment of dyslipidemia in kidney transplantation
.
Expert Opin Drug Saf
.
2020
;
19
(
3
):
257
67
.
29.
Luk
AJ
,
Simkin
PA
.
Epidemiology of hyperuricemia and gout
.
Am J Manag Care
.
2005
;
11
(
15 Suppl
):
S435
42
; quiz S465-8.
30.
Zhou
Z
,
Dong
Y
,
Zhou
H
,
Liu
J
,
Zhao
W
.
Mir-143-3p directly targets glut9 to reduce uric acid reabsorption and inflammatory response of renal tubular epithelial cells
.
Biochem Biophys Res Commun
.
2019
;
517
(
3
):
413
20
.
31.
Bellomo
G
.
Asymptomatic hyperuricemia following renal transplantation
.
World J Nephrol
.
2015
;
4
(
3
):
324
9
.
32.
Pugliese
NR
,
Mengozzi
A
,
Virdis
A
,
Casiglia
E
,
Tikhonoff
V
,
Cicero
AFG
, et al
.
The importance of including uric acid in the definition of metabolic syndrome when assessing the mortality risk
.
Clin Res Cardiol
.
2021
;
110
(
7
):
1073
82
.
33.
Lamouille
S
,
Xu
J
,
Derynck
R
.
Molecular mechanisms of epithelial-mesenchymal transition
.
Nat Rev Mol Cell Biol
.
2014
;
15
(
3
):
178
96
.
34.
Xiong
XY
,
Bai
L
,
Bai
SJ
,
Wang
YK
,
Ji
T
.
Uric acid induced epithelialmesenchymal transition of renal tubular cells through pi3k/p-akt signaling pathway
.
J Cell Physiol
.
2019
;
234
(
9
):
15563
9
.
35.
Zhou
Y
,
Fang
L
,
Jiang
L
,
Wen
P
,
Cao
H
,
He
W
, et al
.
Uric acid induces renal inflammation via activating tubular NF-κb signaling pathway
.
PLoS One
.
2012
;
7
(
6
):
e39738
.
36.
Sharif
A
,
Hecking
M
,
de Vries
AP
,
Porrini
E
,
Hornum
M
,
Rasoul-Rockenschaub
S
, et al
.
Proceedings from an international consensus meeting on posttransplantation diabetes mellitus: recommendations and future directions
.
Am J Transplant
.
2015
;
14
:
1992
2000
.
37.
Ghisdal
L
,
Van Laecke
S
,
Abramowicz
MJ
,
Vanholder
R
,
Abramowicz
D
.
New-onset diabetes after renal transplantation: risk assessment and management
.
Diabetes Care
.
2012
;
35
(
1
):
181
8
,
38.
Ahmed
SH
,
Biddle
K
,
Augustine
T
,
Azmi
S
.
Post-transplantation diabetes mellitus
.
Diabetes Ther
.
2020
;
11
(
4
):
779
801
.
39.
Rodríguez-Rodríguez
AE
,
Porrini
E
,
Hornum
M
,
Donate-Correa
J
,
Morales-Febles
R
,
Khemlani Ramchand
S
, et al
.
Post-transplant diabetes mellitus and prediabetes in renal transplant recipients: an update
.
Nephron
.
2021
;
145
(
4
):
317
29
.
40.
Warburg
OA
.
On the origin of cancer cells
.
Science
.
1956
;
123
(
3191
):
309
14
.
41.
Shin
B
,
Benavides
GA
,
Geng
J
,
Koralov
SB
,
Hu
H
,
Darley-Usmar
VM
, et al
.
Mitochondrial oxidative phosphorylation regulates the fate decision between pathogenic th17 and regulatory t cells
.
Cell Rep
.
2020
;
30
(
6
):
1898
909.e4
.
42.
Ma
EH
,
Verway
MJ
,
Johnson
RM
,
Roy
DG
,
Steadman
M
,
Hayes
S
, et al
.
Metabolic profiling using stable isotope tracing reveals distinct patterns of glucose utilization by physiologically activated cd8+ t cells
.
Immunity
.
2019
;
51
(
5
):
856
70.e5
.
43.
Howden
AJM
,
Hukelmann
JL
,
Brenes
A
,
Spinelli
L
,
Sinclair
LV
,
Lamond
AI
, et al
.
Quantitative analysis of t cell proteomes and environmental sensors during t cell differentiation
.
Nat Immunol
.
2019
;
20
(
11
):
1542
54
.
44.
Carr
EL
,
Kelman
A
,
Wu
GS
,
Gopaul
R
,
Senkevitch
E
,
Aghvanyan
A
, et al
.
Glutamine uptake and metabolism are coordinately regulated by ERK/MAPK during t lymphocyte activation
.
J Immunol
.
2010
;
185
(
2
):
1037
44
.
45.
Nabe
S
,
Yamada
T
,
Suzuki
J
,
Toriyama
K
,
Yasuoka
T
,
Kuwahara
M
, et al
.
Reinforce the antitumor activity of cd8+ t cells via glutamine restriction
.
Cancer Sci
.
2018
;
109
(
12
):
3737
50
.
46.
Wang
R
,
Dillon
CP
,
Shi
LZ
,
Milasta
S
,
Carter
R
,
Finkelstein
D
, et al
.
The transcription factor myc controls metabolic reprogramming upon t lymphocyte activation
.
Immunity
.
2011
;
35
(
6
):
871
82
.
47.
Martínez-Reyes
I
,
Chandel
NS
.
Mitochondrial TCA cycle metabolites control physiology and disease
.
Nat Commun
.
2020
;
11
(
1
):
102
.
48.
Blagih
J
,
Coulombe
F
,
Vincent
EE
,
Dupuy
F
,
Galicia-Vázquez
G
,
Yurchenko
E
, et al
.
The energy sensor ampk regulates t cell metabolic adaptation and effector responses in vivo
.
Immunity
.
2015
;
42
(
1
):
41
54
.
49.
Johnson
MO
,
Wolf
MM
,
Madden
MZ
,
Andrejeva
G
,
Sugiura
A
,
Contreras
DC
, et al
.
Distinct regulation of th17 and th1 cell differentiation by glutaminase-dependent metabolism
.
Cell
.
2018
;
175
(
7
):
1780
95.e19
.
50.
Sinclair
LV
,
Howden
AJ
,
Brenes
A
,
Spinelli
L
,
Hukelmann
JL
,
Macintyre
AN
, et al
.
Antigen receptor control of methionine metabolism in t cells
.
Elife
.
2019
;
8
:
e44210
.
51.
Lu
SC
.
S-adenosylmethionine
.
Int J Biochem Cell Biol
.
2000
;
32
:
391
5
.
52.
Ma
EH
,
Bantug
G
,
Griss
T
,
Condotta
S
,
Johnson
RM
,
Samborska
B
, et al
.
Serine is an essential metabolite for effector t cell expansion
.
Cell Metab
.
2017
;
25
(
2
):
345
57
.
53.
Ron-Harel
N
,
Santos
D
,
Ghergurovich
JM
,
Sage
PT
,
Reddy
A
,
Lovitch
SB
, et al
.
Mitochondrial biogenesis and proteome remodeling promote one-carbon metabolism for t cell activation
.
Cell Metab
.
2016
;
24
(
1
):
104
17
.
54.
Roy
DG
,
Chen
J
,
Mamane
V
,
Ma
EH
,
Muhire
BM
,
Sheldon
RD
, et al
.
Methionine metabolism shapes T helper cell responses through regulation of epigenetic reprogramming
.
Cell Metab
.
2020
;
31
(
2
):
250
66.e9
.
55.
Geiger
R
,
Rieckmann
JC
,
Wolf
T
,
Basso
C
,
Feng
Y
,
Fuhrer
T
, et al
.
L-arginine modulates t cell metabolism and enhances survival and anti-tumor activity
.
Cell
.
2016
;
167
(
3
):
829
42.e13
.
56.
Munn
DH
,
Sharma
MD
,
Baban
B
,
Harding
HP
,
Zhang
Y
,
Ron
D
, et al
.
Gcn2 kinase in t cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase
.
Immunity
.
2005
;
22
(
5
):
633
42
.
57.
Ron-Harel
N
,
Ghergurovich
JM
,
Notarangelo
G
,
Lafleur
MW
,
Tsubosaka
Y
,
Sharpe
AH
, et al
.
T cell activation depends on extracellular alanine
.
Cell Rep
.
2019
;
28
(
12
):
3011
21.e4
.
58.
Sharif
A
,
Ravindran
V
,
Dunseath
G
,
Luzio
S
,
Owens
DR
,
Baboolal
K
:
Comparison of rival metabolic syndrome classifications against pathophysiological markers in renal transplant recipients
.
Transplantation
.
2010
;
89
:
347
52
.
59.
Luan
FL
,
Langewisch
E
,
Ojo
A
.
Metabolic syndrome and new onset diabetes after transplantation in kidney transplant recipients
.
Clin Transplantation
.
2010
;
24
:
778
83
.
60.
Hami
M
,
Sabbagh
M
,
Sefidgaran
A
,
Mojahedi
MJ
.
Prevalence of the metabolic syndrome in kidney transplant recipients: A single-center study
.
Saudi J Kidney Dis Transpl
.
2017
;
28
:
362
7
.
61.
Lee
YH
,
Song
SH
,
Song
SH
,
Shin
HS
,
Yang
J
,
Kim
MS
, et al
.
Clinical implications of changes in metabolic syndrome status after kidney transplantation: a nationwide prospective cohort study
.
Nephrol Dial Transplant
.
2023
;
38
:
2743
53
.
62.
Korogiannou
M
,
Sarafidis
P
,
Theodorakopoulou
MP
,
Alexandrou
M-E
,
Xagas
E
,
Boletis
IN
, et al
.
Diagnostic performance of office versus ambulatory blood pressure in kidney transplant recipients
.
Am J Nephrol
.
2021
;
52
:
548
58
.
63.
Arabi
Z
,
Fawzy
N
,
Arabi
T
,
Sabbah
B
,
Alhamzah
H
,
Alhejaili
F
.
Incidence and factors associated with uncontrolled hypertension at 12 months after kidney transplantation in saudi arabia
.
Kidney Blood Press Res
.
2023
;
48
:
476
84
.
64.
Speer
C
,
Benning
L
,
Morath
C
,
Zeier
M
,
Frey
N
,
Opelz
G
, et al
.
Blood pressure goals and outcomes in kidney transplant recipients in an analysis of the collaborative transplant study
.
Kidney Int Rep
.
2025
;
10
(
3
):
780
90
.
65.
Numakura
K
,
Kagaya
H
,
Yamamoto
R
,
Komine
N
,
Saito
M
,
Hiroshi
T
, et al
.
Characterization of clinical and genetic risk factors associated with dyslipidemia after kidney transplantation
.
Dis Markers
.
2015
;
2015
:
179434
.
66.
Borda
B
,
Szederkényi
E
,
Lengyel
C
,
Morvay
Z
,
Eller
J
,
Marofka
F
, et al
.
Functional and histopathologic changes in renal transplant patients with new-onset diabetes and dyslipidemia
.
Transplant Proc
.
2011
;
43
(
4
):
1254
8
.
67.
Joss
N
,
Staatz
CE
,
Thomson
AH
,
Jardine
AG
.
Predictors of new onset diabetes after renal transplantation
.
Clin Transplant
.
2007
;
21
:
136
43
.
68.
Munshi
VN
,
Saghafian
S
,
Cook
CB
,
Werner
KT
,
Chakkera
HA
.
Comparison of post-transplantation diabetes mellitus incidence and risk factors between kidney and liver transplantation patients
.
PLoS One
.
2020
;
15
:
e0226873
.
69.
Schweer
T
,
Gwinner
W
,
Scheffner
I
,
Schwarz
A
,
Haller
H
,
Blume
C
.
High impact of rejection therapy on the incidence of post‐transplant diabetes mellitus after kidney transplantation
.
Clin Transplant
.
2014
;
28
:
512
9
.
70.
Paek
JH
,
Kang
SS
,
Park
WY
,
Jin
K
,
Park
SB
,
Han
S
, et al
.
Incidence of post-transplantation diabetes mellitus within 1 year after kidney transplantation and related factors in korean cohort study
.
Transplant Proc
.
2019
;
51
:
2714
7
.
71.
Du
Q
,
Li
T
,
Yi
X
,
Song
S
,
Kang
J
,
Jiang
Y
.
Prevalence of new-onset diabetes mellitus after kidney transplantation: A systematic review and meta-analysis
.
Acta Diabetol
.
2024
;
61
:
809
29
.
72.
Mazzali
M
,
Sesti
G
,
Rivelli
G
.
Post transplant hyperuricemia: Incidence and risk factors
.
Transplantation
.
2014
;
98
:
521
.