Next Article in Journal
Comparative Efficacy of Interventions for the Management of Oral Submucous Fibrosis: A Systematic Review and Network Meta-Analysis
Next Article in Special Issue
Delayed Graft Function in Kidney Transplant: Risk Factors, Consequences and Prevention Strategies
Previous Article in Journal
Wnt/β-Catenin-Pathway Alterations and Homologous Recombination Deficiency in Cholangiocarcinoma Cell Lines and Clinical Samples: Towards Specific Vulnerabilities
Previous Article in Special Issue
Cardiovascular Risk after Kidney Transplantation: Causes and Current Approaches to a Relevant Burden
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Non-Immunologic Causes of Late Death-Censored Kidney Graft Failure: A Personalized Approach

by
Claudio Ponticelli
1 and
Franco Citterio
2,*
1
Independent Researcher, Via Ampere 126, 20131 Milano, Italy
2
Renal Transplant Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica Sacro Cuore, 00168 Roma, Italy
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2022, 12(8), 1271; https://doi.org/10.3390/jpm12081271
Submission received: 1 April 2022 / Revised: 28 July 2022 / Accepted: 28 July 2022 / Published: 1 August 2022
(This article belongs to the Special Issue Personalized Medicine in Kidney Transplantation and Immunology)

Abstract

:
Despite continuous advances in surgical and immunosuppressive protocols, the long-term survival of transplanted kidneys is still far from being satisfactory. Antibody-mediated rejection, recurrent autoimmune diseases, and death with functioning graft are the most frequent causes of late-kidney allograft failure. However, in addition to these complications, a number of other non-immunologic events may impair the function of transplanted kidneys and directly or indirectly lead to their failure. In this narrative review, we will list and discuss the most important nonimmune causes of late death-censored kidney graft failure, including quality of the donated kidney, adherence to prescriptions, drug toxicities, arterial hypertension, dyslipidemia, new onset diabetes mellitus, hyperuricemia, and lifestyle of the renal transplant recipient. For each of these risk factors, we will report the etiopathogenesis and the potential consequences on graft function, keeping in mind that in many cases, two or more risk factors may negatively interact together.

1. Introduction

Today, kidney transplantation is the preferred treatment for patients with end-stage kidney disease. Declining rates of acute rejection have led to improvements in short-term kidney transplant survival, but long-term results are still far from being satisfactory. Apart from death with functioning graft, chronic antibody-mediated rejection and recurrence of autoimmune diseases are the major limits for long-term graft survival [1,2,3,4,5]. For this reason, physicians’ attention in the follow-up of transplant recipients is generally focused on immune factors, whereas nonimmunologic factors receive less attention, despite their substantially high contribution to post-transplant morbidity.
In this paper, we will review the most important nonimmunologic causes of death-censored kidney allograft failure.

1.1. Quality of the Donated Kidney

Renal function in renal transplant recipients (RTRs) in general is supported by a solitary functioning kidney. In the case of a deceased donation, the estimated glomerular filtration (eGFR) of the donor usually ranges between 60 and 100 mL/min/1.73 m2, but in older and extended criteria donors (ECDs) the eGFR may be considerably lower. Although any functioning kidney transplant is cost-effective, compared to remaining on dialysis on a waitlist [6], there is evidence that subclinical functional defects are generated in a solitary kidney leading to long-term progressive vasculo-glomerular and tubulo-interstitial lesions, also in nontransplanted individuals [7,8]. Early after transplantation, brain death cytokine storm, ischemia-reperfusion injury, acute rejection, and infections [9,10,11,12,13,14] may lead to acute kidney injury (AKI), ending in a chronic or acute inflammatory state concurring in the pathogenesis of arterial hypertension [15,16]. Another major consequence of the inflammatory state is the development of chronic hypoxia that through the mediation of interleukine 1 and 6, angiotensin II, and transforming growth factor beta can result in excessive accumulation of extracellular matrix and fibrosis [17,18,19]. The excessive deposit of the extracellular matrix can induce epithelial-mesenchymal transition and lead to interstitial fibrosis and chronic allograft dysfunction [20,21,22].
These complications are more frequent and severe in transplants from deceased donors, particularly ECDs. Extended criteria donors nowadays are a main source for kidney transplantation. Survival and quality of life are better for ECD kidney transplants in comparison with dialysis at any age [23,24,25]. However, the number of nephron and GFR progressively decreases after the age of 29 [26], favoring the development of hypertension and CKD in the general population. In kidney transplantation, large series reported that a higher age of donor is related to a less kidney survival at 5 years [27]. For this reason, organs from elderly donors are mainly allocated to elderly recipients, unless the older donor has no history of hypertension, no increased creatinine, no cerebrovascular death, no diabetes or cancer, and no other reasons to be defined as a marginal donor [28].
Living donor transplants in general are followed by a lower release of pro-inflammatory factors in comparison with a deceased donor transplant. Cold ischemia time, ischemia-reperfusion injury, cytokine and autonomic storms related to brain death, are considerably reduced in living donor transplants. Preemptive transplantation also reduces the proinflammatory status, mainly linked to dialysis [29,30].

1.2. Adherence to Prescriptions

Medication non-adherence is prevalent in around 39% of patients with CKD and is associated with poor control of blood pressure, disease progression, adverse events, and mortality [31]. In RTRs the major problem is the complexity of treatment even in patients with stable graft function. The burden of pills that patients receive is not only due to immunosuppressive regimens but also to hypertension, dyslipidemia, and anemia. In a survey of 1130 RTRs with stable serum creatinine, an average 5.9 years after transplantation, each patient was taking on average 11 pills per day, 6 immunosuppressant and 5 non-immunosuppressant [32]. Many immunosuppressive agents used in organ transplantation exert adverse side effects including gastrointestinal disorders, hyperglycemia, hypertension, and possible neurologic complications. Another issue that transplant physicians should be aware of is depression, which is present in 25% of RTRs [33,34]. The presence of depressive symptoms negatively affects adherence to prescriptions and clinical outcomes. Poor adherence is particularly frequent in adolescents, with a weighted prevalence of non-adherence in 32% of RTRs younger than 21 years [35]. In adult RTRs non-adherence to immunosuppressive medications is common, with 20% to 55% of patients being non-adherent [36,37,38]. Poor adherence is often related to social isolation, low belief in medications and/or poor socio-economic conditions. Unintentional compliance is frequent in older patients. They often forget to take medications, and the greater the number of drugs to take, the higher the risk of forgetfulness. Moreover, changes in the dosage or type of medications can often be misunderstood. A systematic review showed that nonadherence was associated with poor clinical outcomes, contributing to 20% of late rejection episodes and 16% of graft losses [39]. Poor adherence to clinical visits and/or to nonimmunosuppressive medications may favor the occurrence of infection, tumors, and CVD. Noncompliance is also favored by the complexity of the immunosuppressive regimen prescribed. Many immunosuppressive drugs are prescribed twice a day, irrespective of available once-a-day formulations and of their pharmacokinetic profile. Studies on patients receiving antibiotics have clearly shown that compliance is strictly related to prescriptions schedules. Morning once-a-day administrations had the best patient compliance. A meta-analysis of randomized controlled trials showed that compliance to antibiotic treatment is higher when an antibiotic is administered once a day than multiple times daily for the treatment of specific infections and for specific classes of antibiotics [40]. Lifetime treatment with immunosuppressive drugs should be personalized and designed, keeping in mind simplicity, to enhance patient compliance.

1.3. Drug Toxicities

Calcineurin inhibitors (CNI) may be responsible for progressive and irreversible kidney toxicities. CNI can cause kidney vasoconstriction by increased expression of vasoconstrictor factors, such as angiotensin II, endothelin-1, thromboxane A2, and leukotrienes, while reducing the production of vasodilators, such as prostacyclin, prostaglandin E2, and nitric oxide. In the long term, a chronic nephrotoxicity may develop, characterized by interstitial fibrosis, tubular atrophy, glomerular sclerosis, and afferent arteriolopathy (Figure 1) [41]. Nephrotoxicity is usually dose-dependent, related to intra- and inter-individual bioavailability and sensitivity. In the 1980s, during first experiences, cyclosporine (CsA) in kidney transplantation was used at elevated doses, up to 15 mg/kg/day. High dosages resulted in frequent acute and chronic nephrotoxicities. An Australian study pointed out that 58% transplanted kidneys treated with CsA for 10 years showed histologic signs of dose-dependent chronic toxicities, such as arteriolar lesions, patchy interstitial fibrosis, tubular atrophy, and focal, segmental, and global glomerular sclerosis [42]. In the same way, when tacrolimus (TAC) was introduced as an immunosuppressive agent, target levels were in the range of 15–20 ng/mL. Slowly, CSA and TAC dosage and blood target levels were considerably reduced. Today, initial doses of CsA and TAC do not exceed 3–5 mg/kg/day and 0.1 mg/kg/day, respectively. Doses are then progressively reduced over the time, up to minimal effective doses for maintenance. To prevent nephrotoxicity, it is also important to avoid concomitant use of nephrotoxic drugs (quinolones, aminoglycosides, amphotericin B, foscarnet, etc.) or drugs that modify the pharmacokinetics of CNI, interfering with cytochrome P 450 activities. The role of CNI as a main cause of interstitial fibrosis has been overevaluated as many authors have suggested [1,43,44]. High CNI exposure can also lead to systemic effects, including arterial hypertension, dyslipidemia, glucose intolerance, hyperuricemia, indirectly affecting kidney allograft function.
An important, still unsolved issue in CNI monitoring is the relationship between current therapeutic drug monitoring based on blood levels and the intracellular concentration of CNI. CNI exert their action inside T lymphocytes. Studies show that blood levels do not reflect the intracellular concentration of CNI and may be misleading [45]. In addition, one should take into account the impact of concurrent drugs that can interfere with CYP450 or P glycoprotein, thus modifying the blood levels [46]. An expert consensus pointed out that acute rejection and kidney toxicity still occur in patients showing blood CNI concentrations within the therapeutic range [47]. Because CNIs exert their action inside T lymphocytes, intracellular CNIs should provide reliable results. However, measuring intracellular CNIs concentrations is not easy. To date, there are no guidelines for the TDM of intracellular CNI concentrations. A recent clinical study in kidney transplantation looked at the relationship between intracellular TAC concentration and rejection or drug-related toxicities. Tacrolimus was measured in the blood and within the cells on days 3 and 10 after kidney transplantation, and on the morning of a for-cause kidney transplant biopsy. The correlation between TAC in the cells and TAC in the blood was poor. The study conclusion was that TAC concentration in cells was not significantly associated with the occurrence of rejection. These results might be inconclusive because of the low number of patients included in this study and because peripheral blood mononuclear cells are not a specific enough matrix to monitor tacrolimus [48]. In the future, the combination of pharmacokinetics, pharmacogenetics, pharmacodynamics, and immunologic biomarkers may allow a better guide the use of TAC in RTRs. The inhibitors of the mammalian target of rapamycine (mTOR), sirolimus, and everolimus can exert dose-dependent antiproliferative and apoptotic effects on epithelial tubular cells [49]. Impaired tubular reabsorption of albumin has been demonstrated in a transplant patient treated with sirolimus in 2006 [50]. A randomized controlled trial comparing CsA with sirolimus reported increased proteinuria, increased urinary excretion of markers of tubular damage and evidence of tubular injury on kidney biopsy in patients treated with sirolimus [51]. Taken together, these data would speak in favor of a tubular toxicity leading to poor reabsorption of albumin and small proteins. Other immunosuppressive drugs used in kidney transplantation do not exert direct nephrotoxicity, including belatacept, purine synthesis inhibitors, proteasome inhibitors, and polyclonal and monoclonal antibodies.

1.4. Dialysis Vintage

End-stage renal disease (ESRD) is a wasting illness and is often associated with a higher risk of cardiovascular diseases and many other comorbidities. A retrospective analysis of the United States Renal Data System Registry reported that compared to preemptive transplantation, waiting times of 0 to 6 months, 6 to 12 months, 12 to 24 months and over 24 months conferred a 17%, 37%, 55%, and 68% increase in risk, respectively, for death-censored graft loss after transplantation [52]. Poor graft function risk is particularly increased in elderly patients [53].

1.5. Hypertension

Arterial hypertension is frequently observed in RTRs and pathogenesis is multifactorial in most cases (Figure 2). Many patients are already hypertensive before transplantation and CNI immunosuppression may induce de novo or worsen previous hypertension. CNI, as a side effect, activates the renin–angiotensin system and deactivates the atrial natriuretic peptide, leading to arteriolar vasoconstriction with consequentially reduced GFR and extracellular fluid expansion. [54]. Additional causes of hypertension are steroid-induced water and salt retention, further aggravated by increased extracellular volume and renin production caused by impaired graft function. Other causes may also contribute to the development of post-transplant hypertension: native kidney disease, old donor age, chronic rejection, and transplant renal artery stenosis. All these factors contribute to extracellular expansion and increase in cardiac output. In the meantime, there is an increase in peripheral vascular resistance caused by inappropriate secretion of renin-angiotensin-aldosterone axis [55].
Arterial hypertension not only increases the risk for cardiovascular events but can also deteriorate kidney allograft function. Several studies have shown that the higher the levels of blood pressure are, the higher is the risk of graft failure [56,57,58,59,60]. Kasiske et al. [61] found that a 10-mmHg increment above 140 mmHg in systolic blood pressure was associated with a 12% relative risk for graft failure and 18% relative risk of death. On the other hand, good blood pressure control may prevent many cardiovascular and kidney complications. Appropriate lifestyle behaviour and physical activity is the first step to control hypertension. Diuretics exert anti-hypertensive effect by reducing salt and water overload but in transplant recipients this may cause a drop in GFR due to the impaired hemodynamic adaptation of the transplanted kidney. Calcium channel blockers reduce systemic vascular resistance acting on vascular smooth cells and may protect one from CNI-induced vasoconstriction. Renin–angiotensin system (RAS) inhibitors control arterial hypertension, reduce proteinuria, and may treat erythrocytosis, but a meta-analysis of three randomized controlled clinical trials and two cohort studies, including 20.024 RTRs, showed no significant reduced risk of allograft loss or mortality in RTRs treated with RAS inhibitors [62]. Current convincing data to prove that RAS inhibitors can actually improve outcomes in RTRs are still lacking. Some transplant physicians are reluctant to prescribe these agents, as potassium can be increased, and hemoglobin and GFR can be reduced. To obtain good control of hypertension and to avoid side effects, most kidney transplant recipients are receiving a synergistic combination of antihypertensive drugs.

1.6. Dyslipidemia

Dyslipidemia is a common complication after kidney transplantation [63,64] and is an important contributor to the high rate of cardiovascular diseases in RTRs, also potentially causing kidney dysfunction. Typically, dislipidemia is observed during the first 3 to 6 months after transplantation, when freedom from dialysis allow a free diet, and may then persist for 10 or more years. The pathogenetic mechanisms for lipid disorders after transplantation are multifactorial: excessive dietary intake of saturated fat and eating disorders, poor physical activity, and sedentary habits are common in RTRs. Many transplant recipients become overweight or obese after transplantation [65] and develop diabetes after transplantation [66]. Moreover, poor post-transplant renal function, leading to CKD stage 3 [67], can favor the development of dyslipidemia [68]. However, the most important cause of post-transplant dyslipidemia is due to immunosuppressive drugs: CSA, TAC, and mTOR inhibitors. Cyclosporine increases total cholesterol (C), VLDL-C, and LDL-C by downregulating LDL receptor expression [69]. Hypertriglyceridemia in CNI-treated RTRs is associated with increased plasma apoCIII concentrations [70]. Despite hypercholesterolemia being significantly less frequent in RTRs receiving TAC with respect to CsA [71], tacrolimus can significantly increase plasma triglycerides, reducing LPL activity [72]. Moreover, mTOR inhibitors are frequently associated with dyslipidemia [73]. These drugs increase hepatic synthesis of apoB100, VLDL, expression of adipose tissue lipase, apoCIII, and lipophagy, while decreasing LDL liver catabolism, LPL expression, and preventing the uptake of lipids into adipocytes [74,75,76]. On the other hand, dyslipidemia caused by mTOR inhibitors may be balanced by the cardioprotective effects of these drugs [67]. Many transplant recipients receive lifetime maintenance glucocorticoids that enhance the activity of acetyl coenzyme convertase and of fatty acid system, increasing hepatic synthesis of VLDL, downregulating LDL receptor activity, and inhibiting lipoprotein lipase [77]. Very few transplant recipients are on a steroid-free regimen. Evidence suggesting that renal lipid accumulation and lipotoxicity may lead to kidney dysfunction has mounted significantly in recent years. Lipid accumulation, changes in circulating adipokines, alterations in renal lipid metabolism, insulin resistance, generation of reactive oxygen species, and endoplasmic reticulum stress are factors eventually leading to damage of the glomerular filtration barrier and to kidney failure [78,79]. Other studies showed that an excessive accumulation of cholesterol and/or triglycerides may cause podocyte injury and proteinuria, suggesting that lipids represent a major regulator of danger signaling from the circulation to glomerular cell [80]. In a retrospective study conducted in more than 12,000 healthy participants, high triglycerides and low HDL-C predicted an increased risk of kidney dysfunction, and treatment of these abnormalities could reduce the incidence of early renal disease [81]. Finally, decreased fatty acid oxidation in CKD may contribute to lipid accumulation in the tubular compartment, which results in energy depletion, followed by apoptosis and de-differentiation, all factors contributing to fibrosis and CKD progression [82]. Although it is difficult to extrapolate these data to RTRs, few studies reported associations between early post-kidney transplant lipid levels and subsequent damages of graft function or death-censored graft loss [83,84]. Pharmacological treatment is necessary if LDL cholesterol level is > 190 mg/dL. Statins are generally well tolerated but myositis and muscle symptoms may occur, leading to poor adherence or discontinuation. There is evidence that statins may reduce cardiovascular events, whereas the benefits on kidney function are controversial. A meta-analysis of randomized controlled trials reported that in adults with CKD, statins do not reduce the risk for kidney failure events, but may modestly reduce proteinuria and rate of eGFR decline [85,86]. Drug–drug interactions that increase statin plasma concentrations mainly involve the co-administration of inhibitors of cytochrome P enzymes (particularly CYP3A4, which is inhibited by CNI), or inhibitors of the transporter proteins activities, which participate in statin cell influx and efflux. Although CNI may increase blood levels of lipophylic statins, with the exception of fluvastatin, results from several studies show that statins do not induce increased systemic exposure of CNI. Ezetimibe represents an alternative, especially for statin-intolerant patients or when added to the highest tolerated statin dose. If a predominant hypertriglyceridemia is present, low calorie intake, low-fat diet and fish oil is suggested and may be beneficial. Fibrates may also lower triglyceride levels but can be responsible for a reversible increase in serum creatinine. There is insufficient information on the use of monoclonal antibodies in RTRs [87].

1.7. New Onset Diabetes Mellitus

New onset diabetes after transplantation (NODAT) is a frequent complication in RTRs, due by traditional and non-traditional pro-diabetic risk factors. The traditional risk factors are the same leading to development of diabetes type 2 in the general population. Non-traditional risk factors include perioperative stress, hepatitis C infection, cytomegalovirus infection, vitamin D deficiency, hypomagnesemia, and immunosuppressive medications, such as glucocorticoids, CNI, and mTOR inhibitors. Glucocorticoids may induce insulin resistance by different mechanisms, including impaired osteoblast function with consequent increase in visceral adiposity and lipolysis, leading to elevated free fatty acids [88], hepatic steatosis [89], and decreased transcription of insulin receptors in skeletal muscle, while increasing transcription of two proteins that counter insulin action [90]. CNI can induce decreased insulin secretion [91], increased insulin resistance [92], and direct toxicity on β cells [93]. The diabetogenic effects of TAC are more intense compared with those of CsA [94]. Immunosuppression with mTOR inhibitors has a direct effect on pancreatic beta cells, reducing directly insulin secretion [95], and inducing gluconeogenic pathways in the liver [96]. However, in RTRs the association between mTOR inhibitors medication and hyperglycemia is weak, and is mostly due to the contemporary drug interaction with CNI [97]. NODAT may progress to diabetic nephropathy and proteinuria. The kidney pathological findings of NODAT are similar to those of primary diabetic nephropathy in native kidneys [98]. However, NODAT is frequently associated with vascular or tubulointerstitial histological changes due to concurrent rejection, viral infections, or drug nephrotoxicity. Avoiding excessive weight gain, leading a healthy lifestyle, reducing caloric intake, and engaging in physical exercise are typically recommended at the discharge of patients after renal transplantation, but compliance with these indications is generally low. Management of diabetes is more challenging in transplant recipients than in nontransplant patients, because of the greater risk on kidney function. Metformin may offer some advantages over other glucose-lowering agents with respect to risk of hypoglycemia [99] and beneficial effects on the kidney [100,101], There has been concern about the risk of life-threatening lactic acidosis. However, many kidney transplant recipients are using metformin without experiencing side effects [102]. Two comprehensive reviews found no evidence of an increased risk of lactic acidosis, using metformin compared to other anti-hyperglycemic treatments [103,104]. Potential side effects of rosiglitazone and pioglitazone include edema, congestive heart failure, and bone fractures [105,106]. Glucagon-like peptide-1 (GLP-1) inhibitors may reduce progression of renal disease in type 2 diabetes [107,108], but these drugs have serious gastrointestinal effects and might increase the risk of pancreatitis and/or tumors [109]. A small study in organ transplant recipients reported that GLP-1 inhibitors are effective and do not affect TAC levels or transplant outcomes in the short term [110]. Inhibitors of dipeptidyl peptidase 4 (DPP-4 inhibitors or gliptins) are generally considered safe, with a low rate of side effects. Concerns about increased risk of cardiovascular events or pancreatic cancer development have not been confirmed [111]. A systematic review of 7 studies reported that DPP-4 inhibitor use in transplant recipients did not result in significant change in eGFR or TAC blood levels [112]. Sodium glucose cotransporter-2 (SGLT2) inhibitors, such as canagliflozin, dapagliflozin, empaglifloziin, sotagliflozin etc., inhibit the reabsorption of glucose in the proximal tubular cells and facilitate glucose excretion in urine. As glucose is excreted, its plasma levels fall, leading to an improvement in all glycemic parameter. SGLT2 inhibition has also been shown to reduce cardiovascular mortality and preserve kidney function in patients with type 2 diabetes [113]. However, urinary tract infections and a slight initial decrease in renal function may limit use of SGLT2 inhibitors [114]. A systematic review and meta-analysis of 8 studies with 132 RTRs with excellent kidney function reported that SGLT-2 inhibitors for treatment of NODAT are effective in lowering glycate hemoglobin, reducing body weight, and preserving kidney function without serious adverse events [115]. In RTRs requiring insulin treatment, the dose and type of insulin prescription should be based on individual patient needs.

1.8. Hyperuricemia

Hyperuricemia is related to cardiovascular diseases [116,117] and is an independent predictor of chronic kidney disease development and progression [118,119,120,121,122]. An independent association between serum urate levels and allograft outcomes was found in RTRs [123,124]. A retrospective analysis in 2993 kidney transplant recipients showed that low and normal serum urate levels within the first year are an independent predictor of better renal allograft outcomes in the long term [125]. However, in RTRs it is difficult to separate hyperuricemia as a cause or consequence of kidney injuries [126]. The mechanistic interpretation of urate-related kidney injury is complex. Hyperuricemia can activate the renin-angiotensin-aldosterone system, along with the inhibition of nitric-oxide synthesis in the kidney, leading to medial thickening of preglomerular arterioles, renal vasoconstriction, and increased systemic blood pressure [127]. On the other hand, uric acid is recognized as an endogenous damage-associated molecular pattern by pattern-recognition receptors with engagement of inflammasomes and activation of pro-inflammatory interleukin-1β and interleukin-18 [128]. This inflammatory process may end in kidney ischemia and hypoxia, two powerful inducers of tubulointerstitial fibrosis [129], and in the epithelial-mesenchymal transition of renal tubular cells with increased fibronectin synthesis [130,131]. In summary, a two-hit model can be proposed. The first hit entails activation of molecules and factors that promote endothelial dysfunction, proliferation of vascular smooth-muscle cells and sodium reabsorption, leading to increased systemic blood pressure. The second hit involves activation of inflammatory status increasing vascular resistances, kidney ischemia and hypoxia, eventually leading to vascular and tubulo-interstitial lesions inducing development and progression of kidney disease [132]. Xanthine oxidase inhibitors are the cornerstone to reduce uricemia to ≤6 mg/dL, the target value for urate-lowering drugs. Allopurinol is a nonspecific competitive inhibitor of xanthine oxidase that undergoes conversion to oxypurinol, prior to renal excretion. Serious adverse events are mainly related to hypersensitivity to allopurinol [133]. Concomitant administration of azathioprine and xanthine oxidase inhibitors engenders the hazard of severe bone marrow suppression because xanthine oxidase inhibitors inhibit the oxidation of 6-mercaptopurine to inactive metabolites. Therefore, concomitant administration should be avoided, unless allopurinol is the only option and has to be used. In these cases, dose reduction of azathioprine or 6-mercaptopurine is suggested, and white blood cells should be strictly monitored. More recently, febuxostat has become an established alternative for the treatment of hyperuricemia. Febuxostat is extensively metabolized by oxidation and acyl-glucuronidation, with subsequent renal clearance of febuxostat-acyl-glucuronides. Although pharmacokinetic parameters are not affected by mild to moderate hepatic impairment, there is no consensus on whether renal impairment has any effect on the pharmacokinetics of febuxostat [134]. As for allopurinol, current febuxostat labeling contraindicates the concomitant administration of febuxostat with either azathioprine or 6-mercaptopurine. In RTR, febuxostat had higher odds to reach the target of serum uric acid < 6 mg/dL compared to allopurinol, without causing significant side effects [135,136].
Although mounting evidence indicate that hyperuricemia may concur with other factors in inducing kidney damages, no robust data are available to support the routine use of pharmacotherapy for RTRs with asymptomatic hyperuricemia.

1.9. Anemia

In transplant recipients with poor kidney allograft function, anemia is frequent. Erythropoiesis stimulating agents (ESAs) may be used in the early posttransplant period to prevent renal hypoxia or in the late period to prevent cardiovascular complications. There is no evidence that treatment with ESAs may affect kidney function in RTRs. Rather, targeting higher hemoglobin levels may reduce the progression of allograft nephropathy [137].

2. Lifestyle

2.1. Smoking

Epidemiological studies documented a marked risk of irreversible proteinuria in smokers [137,138]. Smoking can also accelerate the progression of renal failure in patients with kidney disease [139]. Patients who continue to smoke after transplantation are at increased risk of graft failure [140] that may be reversed by stopping smoking [141]. A history of smoking before kidney transplantation can also contribute significantly to allograft loss [142,143]. The kidney histologic picture in smokers is characterized by interstitial fibrosis [144] or nodular glomerulosclerosis [145]. The pathogenesis of smoking-related renal damage is largely unknown. The intermittent increase in blood pressure during smoking might play a major role in causing renal damage [146]. Increased sympathetic activity, increased renal vascular resistance, oxidative stress, increased intraglomerular pressure, and/or renal artery arteriosclerosis probably contributes to the deleterious effects of smoking [147].

2.2. Sedentary Activity

A retrospective study evaluated the impact on kidney function of active physical activity (≥ 30-min, 5 times/week) versus nonactive patients in 2060 stable RTRs aged ≥18 years, with at least a 10-year follow-up. A slower decline of eGFR over time was observed in active RTRs compared to non-active patients [148]. It is unclear how physical activity may protect one from kidney function deterioration. There is evidence that excessive weight, glucose intolerance, and high blood pressure are reduced by regular physical activities, indirectly avoiding eGFR decline [149]. In a recent paper, 19 RTRs prospectively underwent magnetic resonance images on a 3T scanner including diffusion-weighted, blood oxygenation level dependent (BOLD), and arterial spin labeling sequences in hip positions 00 and >900 before and after intravenous administration of 20 mg furosemide. Unexpectedly, graft perfusion values were significantly higher in flexed, compared to neutral hip position. BOLD-derived cortico-medullary R2 ratio was significantly modified during hip flexion, suggesting an intrarenal redistribution of the oxygenation in favor of the medulla and to the detriment of the cortex. Although more data are needed, the authors suggest avoiding prolonged sitting for RTRs and favor exercises without major sustained hip flexion, such as rowing, for example [150].

2.3. Diet

Nutrition is an important medical aspect in kidney transplantation. During the acute post-transplant phase, some patients with a long history of CKD and dialysis are generally malnourished [151,152]. In these patients, dietetic regimen of 25–35 kcal/kg ideal body weight (IBW)/day and 1.0–1.2 g protein/kg IBW/day is recommended, until achievement of nutritional adequacy [153,154].
In the long-term, a low-salt and high-fiber diet is recommended to prevent and treat obesity, diabetes, dyslipidemia, and hypertension. The Mediterranean diet exerts beneficial effects on CVD, diabetes, obesity, metabolic syndrome, and cancer [155,156,157,158,159,160]. The Mediterranean diet, including high consumption of olive oil, legumes, unrefined cereals, fruits, and vegetables, a moderate to high consumption of fish, a moderate consumption of dairy products (mostly as cheese and yogurt), a moderate wine consumption, and a low consumption of non-fish meat products, might also protect kidney graft function. In a study, the nine-point Mediterranean Diet Score was assessed in 632 RTRs with graft functioning for ≥1 year, adherent to the Mediterranean diet. During median follow-up of 5.4 years, the Mediterranean Diet Score was inversely associated with graft failure, kidney function decline, and graft loss, independently of potential confounders [161]. For people unfamiliar with the Mediterranean diet, this diet is based on a balanced diet of a variety of fresh fruits and vegetables, lean meats, and plenty of water, whereas fat dairy products and whole grains should be reduced.

2.4. Infections

Any severe infection can damage the kidney graft either because of rejection due to reduced immunosuppression or because of treatment with nephrotoxic anti-infective drugs such as aminoglycosides, vancomycin, amphoterin, quinolones, anti-fungal, and anti-viral agents. Urinary tract infections (UTI) are frequent but usually respond well to antimicrobials or do not need treatment if asymptomatic. However, frequent recurrence of UTI may be associated to an increased risk of chronic rejection [162]. UTI may induce acute pyelonephritis, particularly in patients with stones, stents, or mechanical obstruction. Rapid increase of serum creatinine is possible in pyelonephritis. Acute kidney damages may be reversible if infection is controlled, but may also cause graft loss and death [163] when bacteremia leads to multi-organ dysfunction, with the kidney frequently involved.
Candidosis, aspergillosis, and mucormycosis may cause severe renal disease usually in the early post-transplant period. BK polyoma virus is the most frequent cause of infective late graft failure. Infection occurs during childhood and remains latent in the tubules. In kidney transplant recipients, the reactivation of BK virus may occur. The diagnosis of BKV is made between 6 and 12 months post-transplantation, more often after treatment of rejection and in patients receiving strong immunosuppression [164,165]. BK virus infection may cause interstitial nephritis with a large number of plasma cells that leads to progressive graft loss. The prevalence of polyomavirus BK nephropathy in RTRs ranges between 3% and 8% [166,167,168]. The presence of viral inclusions, known as “decoy cells,” in urine and the presence of BK virus DNA in plasma and in urine are markers for the replication of BK virus infection [166,167]. The sensitivity of decoy cells is 100% but the predictive value is low, only 27%. The demonstration of viremia confirm the diagnosis, with a sensitivity of 100%, a specificity of 92% and a predictive value of 74% [169]. Renal biopsy shows interstitial nephritis with infiltrate rich in plasma cells, and atypical intra-nuclear viral inclusion bodies. Ureteral stenosis and/or hemorrhagic cystitis can also occur. In the presence of renal function, deterioration and positive PCR-BKV renal biopsy is mandatory. There is no specific treatment for BK virus infection. Reduction of immunosuppression may stabilize renal function in a few cases but may expose one to the risk of acute rejection. A systematic review showed that leflunomide, cidofovir, and intravenous immunoglobulins failed to show any efficacy [170]. Graft loss occurs in 15–50% of BK polyomavirus-associated nephropathy. The Transform study has recently shown that everolimus, in combination with CNI, has a protective effect on BKV infection development. If a patient has lost the transplanted kidney because of BKV nephropathy, in the absence of BKV viremia, retransplantation is not a contraindication [171], and the immunosuppressive regimen including everolimus may be protective from reinfection.
In conclusion, today many kidney transplant are lost because of death with functioning graft (47%), chronic allograft nephropathy (29%), acute rejection (2.8%), hyperacute rejection (0.1%), nonlethal cardiovascular events (2%), technical complications (0.8%), recurrent glomerulonephritis (3.2%), noncompliance (2%), and other causes (11.8%) [172].
A personalized approach to care for clinical signs leading to nonimmune kidney graft failure, including adherence to prescriptions, avoiding drug toxicities, controlling arterial hypertension, treating dyslipidaemia, caring for new onset diabetes mellitus, treating hyperuricemia, and suggesting a healthy lifestyle may contribute to prolonged renal transplant function, and give better and longer lives to transplant recipients.

Author Contributions

Conceptualization, C.P., Data creation C.P. and F.C., Writing, C.P. and F.C. Review editing, F.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The study did not report any data.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sellarés, J.; Freitas, D.G.; Mengel, M.; Reeve, J.; Einecke, G.; Sis, B.; Hidalgo, L.G.; Famulski, K.; Matas, A.; Halloran, P.F. Understanding the causes of kidney transplant failure: The dominant role of antibody-mediated rejection and nonadherence. Am. J. Transplant. 2012, 12, 388–399. [Google Scholar] [CrossRef]
  2. Moreso, F.; Carrera, M.; Goma, M.; Hueso, M.; Sellares, J.; Martorell, J.; Grinyó, J.M.; Serón, D. Early subclinical rejection as a risk factor for late chronic humoral rejection. Transplantation 2012, 93, 41–46. [Google Scholar] [CrossRef]
  3. Halloran, P.F.; Reeve, J.P.; Pereira, A.B.; Hidalgo, L.G.; Famulski, K.S. Antibody-mediated rejection, T cell-mediated rejection, and the injury-repair response: New insights from the Genome Canada studies of kidney transplant biopsies. Kidney Int. 2014, 85, 258–264. [Google Scholar] [CrossRef] [Green Version]
  4. Stites, E.; Le Quintrec, M.; Thurman, J.M. The Complement System and Antibody Mediated Transplant Rejection. J. Immunol. 2015, 195, 5525–5531. [Google Scholar] [CrossRef] [Green Version]
  5. Einecke, G.; Reeve, J.; Gupta, G.; Böhmig, G.A.; Eskandary, F.; Bromberg, J.S.; Budde, K.; Halloran, P.F.; INTERCOMEX Investigators. Factors associated with kidney graft survival in pure antibody-mediated rejection at the time of indication biopsy: Importance of parenchymal injury but not disease activity. Am. J. Transpl. 2021, 21, 1391–1401. [Google Scholar] [CrossRef]
  6. Senanayake, S.; Graves, N.; Healy, H.; Baboolal, K.; Barnett, A.; Sypek, M.P.; Kularatna, S. Donor Kidney Quality and Transplant Outcome: An Economic Evaluation of Contemporary Practice. Value Health 2020, 23, 1561–1569. [Google Scholar] [CrossRef]
  7. Sanna-Cherchi, S.; Ravani, P.; Corbani, V.; Parodi, S.; Haupt, R.; Piaggio, G.; Innocenti, M.L.; Somenzi, D.; Trivelli, A.; Caridi, G.; et al. Renal outcome in patients with congenital anomalies of the kidney and urinary tract. Kidney Int. 2009, 76, 528–533. [Google Scholar] [CrossRef] [Green Version]
  8. Kim, S.; Chang, Y.; Lee, Y.R.; Jung, H.S.; Hyun, Y.Y.; Lee, K.B.; Joo, K.J.; Yun, K.E.; Shin, H.; Ryu, S. Solitary kidney and risk of chronic kidney disease. Eur. J. Epidemiol. 2019, 34, 879–888. [Google Scholar] [CrossRef] [PubMed]
  9. De Vries, D.K.; Lindeman, J.H.N.; Ringers, J.; Reinders, M.E.J.; Rabelink, T.J.; Schaapherder, A.F.M. Donor Brain Death Predisposes Human Kidney Grafts to a Proinflammatory Reaction after Transplantation. Am. J. Transpl. 2011, 11, 1064–1070. [Google Scholar] [CrossRef]
  10. Debout, A.; Foucher, Y.; Trébern-Launay, K.; Legendre, C.; Kreis, H.; Mourad, G.; Garrigue, V.; Morelon, E.; Buron, F.; Rostaing, L.; et al. Each additional hour of cold ischemia time significantly increases the risk of graft failure and mortality following renal transplantation. Kidney Int. 2015, 87, 243–249. [Google Scholar] [CrossRef] [Green Version]
  11. Thurman, J.M. Triggers of inflammation after renal ischemia/reperfusion. Clin. Immunol. 2007, 123, 7–13. [Google Scholar] [CrossRef] [Green Version]
  12. Ponticelli, C. The impact of cold ischemia time on renal transplant outcome. Kidney Int. 2015, 87, 272–275. [Google Scholar] [CrossRef] [Green Version]
  13. Chu, Z.; Sun, C.; Sun, L.; Feng, C.; Yang, F.; Xu, Y.; Zhao, Y. Primed macrophages directly and specifically reject allografts. Cell Mol. Immunol. 2020, 17, 237–246. [Google Scholar] [CrossRef]
  14. Mori, D.N.; Kreisel, D.; Fullerton, J.N.; Gilroy, D.W.; Goldstein, D.R. Inflammatory triggers of acute rejection of organ allografts. Immunol. Rev. 2014, 258, 132–144. [Google Scholar] [CrossRef] [Green Version]
  15. Dinh, Q.N.; Drummond, G.R.; Sobey, C.G.; Chrissobolis, S. Roles of inflammation, oxidative stress, and vascular dysfunction in hypertension. Biomed. Res. Int. 2014, 2014, 406960. [Google Scholar] [CrossRef] [Green Version]
  16. Xiao, L.; Harrison, D.G. Inflammation in Hypertension. Can. J. Cardiol. 2020, 36, 635–647. [Google Scholar] [CrossRef] [PubMed]
  17. Yamaguchi, J.; Tanaka, T.; Eto, N.; Nangaku, M. Inflammation and hypoxia linked to renal injury by CCAAT/enhancer-binding protein. Kidney Int. 2015, 88, 262–275. [Google Scholar] [CrossRef] [Green Version]
  18. Burns, K.D. Interleukin-1beta as a mediator of tubulointerstitial fibrosis. Kidney Int. 2002, 62, 346–347. [Google Scholar] [CrossRef] [Green Version]
  19. Jordan, S.C.; Choi, J.; Kim, I.; Wu, G.; Toyoda, M.; Shin, B.; Vo, A. Interleukin-6, A Cytokine Critical to Mediation of Inflammation, Autoimmunity and Allograft Rejection: Therapeutic Implications of IL-6 Receptor Blockade. Transplantation 2017, 101, 32–44. [Google Scholar] [CrossRef]
  20. Tonelli, M.; Sacks, F.; Pfeffer, M.; Jhangri, G.S.; Curhan, G. Biomarkers of inflammation and progression of chronic kidney disease. Kidney Int. 2005, 68, 237–245. [Google Scholar] [CrossRef] [Green Version]
  21. Liu, M.; Ning, X.; Li, R.; Yang, Z.; Yang, X.; Sun, S.; Qian, Q. Signalling pathways involved in hypoxia-induced renal fibrosis. J. Cell Mol. Med. 2017, 21, 1248–1259. [Google Scholar] [CrossRef] [PubMed]
  22. Ponticelli, C.; Campise, R. The inflammatory state is a risk factor for cardiovascular disease and graft fibrosis in kidney transplantation. Kidney Int. 2021, 100, 536–555. [Google Scholar] [CrossRef] [PubMed]
  23. Molnar, M.Z.; Streja, E.; Kovesdy, C.P.; Shah, A.; Huang, E.; Bunnapradist, S.; Krishnan, M.; Kopple, J.D.; Kalantar-Zadeh, K. Age and the associations of living donor and expanded criteria donor kidneys with kidney transplant outcomes. Am. J. Kidney Dis. 2012, 59, 841–848. [Google Scholar] [CrossRef] [Green Version]
  24. Ponticelli, C.; Podestà, M.A.; Graziani, G. Renal transplantation in elderly patients. How to select the candidates to the waiting list? Transpl. Rev. 2014, 28, 188–192. [Google Scholar] [CrossRef] [PubMed]
  25. Gillen, D.L.; Stehman-Breen, C.O.; Smith, J.M.; McDonald, R.A.; Warady, B.A.; Brandt, J.R.; Wong, C.S. Survival advantage of pediatric recipients of a first kidney transplant among children awaiting kidney transplantation. Am. J. Transpl. 2008, 8, 2600–2606. [Google Scholar] [CrossRef] [PubMed]
  26. Denic, A.; Mathew, J.; Nagineni, V.V.; Thompson, R.H.; Leibovich, B.C.; Lerman, L.O.; Lieske, J.C.; Alexander, M.P.; Augustine, J.J.; Kremers, W.K.; et al. Clinical and Pathology Findings Associate Consistently with Larger Glomerular Volume. J. Am. Soc. Nephrol. 2018, 29, 1960–1969. [Google Scholar] [CrossRef] [Green Version]
  27. Echterdiek, F.; Schwenger, V.; Döhler, B.; Latus, J.; Kitterer, D.; Heemann, U.; Süsal, C. Kidneys from Elderly Deceased Donors-Is 70 the New 60? Front. Immunol. 2019, 10, 2701. [Google Scholar] [CrossRef]
  28. Süsal, C.; Kumru, G.; Döhler, B.; Morath, C.; Baas, M.; Lutz, J.; Unterrainer, C.; Arns, W.; Aubert, O.; Bara, C.; et al. Should kidney allografts from old donors be allocated only to old recipients? Transpl. Int. 2020, 33, 849–857. [Google Scholar] [CrossRef] [PubMed]
  29. Ya, Q.; Lindholm, B.; Stenkivel, P. Inflammation as a cause of malnutrition, atherosclerotic, cardiovascular disease and poor outcome in hemodialysis patients. Hemodial. Int. 2004, 29, 118–129. [Google Scholar] [CrossRef] [PubMed]
  30. Li, P.K.; Ng, J.K.; Mcintyre, C.W. Inflammation and Peritoneal Dialysis. Semin. Nephrol. 2017, 37, 54–65. [Google Scholar] [CrossRef]
  31. Tesfaye, W.H.; Erku, D.; Mekonnen, A.; Tefera, Y.G.; Castelino, R.; Sud, K.; Thomas, J.; Obamiro, K. Medication non-adherence in chronic kidney disease: A mixed-methods review and synthesis using the theoretical domains framework and the behavioural change wheel. J. Nephrol. 2021, 34, 1091–1125. [Google Scholar] [CrossRef] [PubMed]
  32. Ponticelli, C.; Colombo, D.; Novara, M.; Basilisco, G. Gastrointestinal symptoms impair quality of life in Italian renal transplant recipients but are under-recognized by physicians. Transpl. Int. 2010, 23, 1126–1134. [Google Scholar] [CrossRef] [PubMed]
  33. Szeifert, L.; Molnar, M.Z.; Ambrus, C.; Koczy, A.B.; Kovacs, A.Z.; Vamos, E.P.; Keszei, A.; Mucsi, I.; Novak, M. Symptoms of depression in kidney transplant recipients: A cross-sectional study. Am. J. Kidney Dis. 2010, 55, 132–140. [Google Scholar] [CrossRef]
  34. Chilcot, J.; Spencer, B.W.; Maple, H.; Mamode, N. Depression and kidney transplantation. Transplantation 2014, 97, 717–721. [Google Scholar] [CrossRef] [PubMed]
  35. Shemesh, E.; Annunziato, R.A.; Arnon, R.; Miloh, T.; Kerkar, N. Adherence to medical recommendations and transition to adult services in pediatric transplant recipients. Curr. Opin. Organ. Transpl. 2010, 15, 288–292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Butler, J.A.; Roderick, P.; Mullee, M.; Mason, J.C.; Peveler, R.C. Frequency and impact of nonadherence to immunosuppressants after renal transplantation: A systematic review. Transplantation 2004, 77, 769–776. [Google Scholar] [CrossRef]
  37. Low, J.K.; Manias, E.; Crawford, K.; Walker, R.; Mulley, W.R.; Toussaint, N.D.; Dooley, M.; Kennedy, E.; Smith, C.L.; Nalder, M.; et al. Improving medication adherence in adult kidney transplantation (IMAKT): A pilot randomised controlled trial. Sci. Rep. 2019, 9, 7734. [Google Scholar] [CrossRef] [Green Version]
  38. Gokoel, S.R.M.; Gombert-Handoko, K.B.; Zwart, T.C.; van der Boog, P.J.M.; Moes, D.J.A.R.; de Fijter, J.W. Medication non-adherence after kidney transplantation: A critical appraisal and systematic review. Transpl. Rev. 2020, 34, 100511. [Google Scholar] [CrossRef]
  39. Denhaerynck, K.; Dobbels, F.; Cleemput, I.; Desmyttere, A.; Schäfer-Keller, P.; Schaub, S.; De Geest, S. Prevalence, consequences, and determinants of nonadherence in adult renal transplant patients: A literature review. Transpl. Int. 2005, 18, 1121–1133. [Google Scholar] [CrossRef]
  40. Falagas, M.E.; Karagiannis, A.K.; Nakouti, T.; Tansarli, G.S. Compliance with once-daily versus twice or thrice-daily administration of antibiotic regimens: A meta-analysis of randomized controlled trials. PLoS ONE 2015, 10, e0116207. [Google Scholar] [CrossRef]
  41. Andoh, F.H.; Bennett, W.M. Chronic cyclosporine nephrotoxicity. Curr. Opin. Nephrol. Hypertens. 1998, 7, 265–270. [Google Scholar] [CrossRef] [PubMed]
  42. Nankivell, B.J.; Borrows, R.J.; Fung, C.L.; O’Connell, P.J.; Allen, R.D.; Chapman, J.R. The natural history of chronic allograft nephropathy. N. Engl. J. Med. 2003, 349, 2326–2333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Venner, J.M.; Famulski, K.S.; Reeve, J.; Chang, J.; Halloran, P.F. Relationships among injury, fibrosis, and time in human kidney transplants. JCI Insight 2016, 1, e85323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Montgomery, R.A.; Loupy, A.; Segev, D. Antibody-mediated rejection: New approaches in prevention and management. Am. J. Transpl. 2018, 18 (Suppl. 3), 3–17. [Google Scholar] [CrossRef] [Green Version]
  45. Naesens, M.; Kuypers, D.R.; Sarwal, M. Calcineurin inhibitor nephrotoxicity. Clin. J. Am. Soc. Nephrol. 2009, 4, 481–508. [Google Scholar] [CrossRef] [Green Version]
  46. Ponticelli, C.; Reggiani, F.; Moroni, G. New and old calcineurin inhibitors in lupus nephritis. J. Clin. Med. 2021, 10, 4832. [Google Scholar] [CrossRef]
  47. Lemaitre, F.; Vethe, N.T.; D’Avolio, A.; Tron, C.; Robertsen, I.; De Winter, B.; Denicolo, A.; Koch, B.C.P.; Venkataramanan, R.; Van Gelder, T.; et al. Measuring Intracellular Concentrations of Calcineurin Inhibitors: Expert Consensus from the International Association of Therapeutic Drug Monitoring and Clinical Toxicology Expert Panel. Ther. Drug Monit. 2020, 42, 665–670. [Google Scholar] [CrossRef]
  48. Francke, M.I.; Andrews, L.M.; Lan Le, H.; van de Velde, D.; Dieterich, M.; Udomkarnjananun, S.; Clahsen-van Groningen, M.C.; Baan, C.C.; van Gelder, T.; de Winter, B.C.M.; et al. Monitoring intracellular tacrolimus concentrations and its relationship with rejection in the early phase after renal transplantation. Clin. Biochem. 2022, 101, 9–15. [Google Scholar] [CrossRef]
  49. Lieberthal, W.; Fuhro, R.; Andry, C.C.; Rennke, H.; Abernathy, V.E.; Koh, J.S.; Valeri, R.; Levine, J.S. Rapamycin impairs recovery from acute renal failure: Role of cell-cycle arrest and apoptosis of tubular cells. Am. J. Physiol. Renal. Physiol. 2001, 281, F693–F706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Straathof-Galema, L.; Wetzels, J.F.; Dijkman, H.B.; Steenbergen, E.J.; Hilbrands, L.B. Sirolimus-associated heavy proteinuria in a renal transplant recipient: Evidence for a tubular mechanism. Am. J. Transpl. 2006, 2, 429–433. [Google Scholar] [CrossRef]
  51. Franz, S.; Regeniter, A.; Hopfer, H.; Mihatsch, M.; Dickenmann, M. Tubular toxicity in sirolimus- and cyclosporine-based transplant immunosuppression strategies: An ancillary study from a randomized controlled trial. Am. J. Kidney Dis. 2010, 55, 335–343. [Google Scholar] [CrossRef] [PubMed]
  52. Meier-Kriesche, H.U.; Port, F.K.; Ojo, A.O.; Rudich, S.M.; Hanson, J.A.; Cibrik, D.M.; Leichtman, A.B.; Kaplan, B. Effect of waiting time on renal transplant outcome. Kidney Int. 2000, 58, 1311–1317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. So, S.; Au, E.H.K.; Lim, W.H.; Lee, V.W.S.; Wong, G. Factors Influencing Long-Term Patient and Allograft Outcomes in Elderly Kidney Transplant Recipients. Kidney Int. Rep. 2021, 6, 727–736. [Google Scholar] [CrossRef] [PubMed]
  54. Curtis, J.J. Hypertension following kidney transplantation. Am. J. Kidney Dis. 1994, 23, 471–475. [Google Scholar] [CrossRef]
  55. Ponticelli, C.; Cucchiari, D.; Graziani, G. Hypertension in kidney transplant recipients. Transpl. Int. 2011, 24, 523–533. [Google Scholar] [CrossRef] [PubMed]
  56. Woo, Y.M.; Pereira, B.J.; Gill, J.S. Chronic kidney disease progression in native and transplant kidneys. Curr. Opin. Nephrol. Hypertens. 2004, 13, 607–611. [Google Scholar] [CrossRef] [PubMed]
  57. Castillo-Lugo, J.A.; Vergne-Marini, P. Hypertension in kidney transplantation. Semin. Nephrol. 2005, 25, 252–260. [Google Scholar] [CrossRef] [PubMed]
  58. Tedla, F.; Hayashi, R.; McFarlane, S.I.; Salifu, M.O. Hypertension after renal transplant. J. Clin. Hypertens. (Greenwich) 2007, 9, 538–545. [Google Scholar] [CrossRef] [PubMed]
  59. Vanrenterghem, Y.F.; Claes, K.; Montagnino, G.; Fieuws, S.; Maes, B.; Villa, M.; Ponticelli, C. Risk factors for cardiovascular events after successful renal transplantation. Transplantation 2008, 85, 209–216. [Google Scholar] [CrossRef]
  60. Vetromile, F.; Szwarc, I.; Garrigue, V.; Delmas, S.; Fesler, P.; Mimran, A.; Ribstein, J.; Mourad, G. Early high pulsepressure is associated with graft dysfunction and predicts poor kidney allograft survival. Transplantation 2009, 8, 1088–1094. [Google Scholar] [CrossRef]
  61. Kasiske, B.L.; Anjum, S.; Shah, R.; Skogen, J.; Kandaswamy, C.; Danielson, B.; O’Shaughnessy, E.A.; Dahl, D.C. Hypertension after kidney transplantation. Am. J. Kidney Dis. 2004, 43, 1071–1081. [Google Scholar] [CrossRef] [PubMed]
  62. Cheungpasitporn, W.; Thongprayoon, C.; Mao, M.A. The effect of renin-angiotensin system inhibitors on kidney allograft survival: A systematic review and meta-analysis. N. Am. J. Med. Sci. 2016, 8, 291–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Andany, M.A.; Kasiske, B.L. Dyslipidemia and its management after renal transplantation. J. Nephrol. 2001, 14 (Suppl. 4), S81–S88. [Google Scholar] [PubMed]
  64. Thobani, A.; Jacobson, T.A. Dyslipidemia in Patients with Kidney Disease. Cardiol. Clin. 2021, 39, 353–363. [Google Scholar] [CrossRef] [PubMed]
  65. Zhang, Y.; Yang, J.; Ye, J.; Guo, Q.; Wang, W.; Sun, Y.; Zeng, Q. Separate and combined associations of physical activity and obesity with lipid-related indices in non-diabetic and diabetic patients. Lipids Health Dis. 2019, 18, 49. [Google Scholar] [CrossRef] [Green Version]
  66. Paoletti, E.; Citterio, F.; Corsini, A.; Potena, L.; Rigotti, P.; Sandrini, S.; Bussalino, E.; Stallone, G.; ENTROPIA Project. Everolimus in kidney transplant recipients at high cardiovascular risk: A narrative review. J. Nephrol. 2019, 33, 69–82. [Google Scholar] [CrossRef]
  67. Costa de Oliveira, C.M.; Mota, M.U.; Mota, R.S.; Nóbrega, J.O.; Melo, D.S.; Vieira, A.S.; Fernandes, P.F.C.B.; Campos, H.d.; Evangelista, J.B. Prevalence and staging of chronic kidney disease in renal transplant recipients. Clin. Transpl. 2009, 23, 628–636. [Google Scholar] [CrossRef]
  68. Kasiske, B.L.; Israni, A.K.; Snyder, J.J.; Skeans, M.A.; Patient Outcomes in Renal Transplantation (PORT) Investigators. The relationship between kidney function and long-term graft survival after kidney transplant. Am. J. Kidney Dis. 2011, 57, 466–475. [Google Scholar] [CrossRef]
  69. Tavori, H.; Rashid, S.; Fazio, S. On the function and homeostasis of PCSK9: Reciprocal interaction with LDLR and additional lipid effects. Atherosclerosis 2015, 238, 264–270. [Google Scholar] [CrossRef] [Green Version]
  70. Badiou, S.; Garrigue, V.; Dupuy, A.M.; Chong, G.; Cristol, J.P.; Mourad, G. Small dense low-density lipoprotein in renal transplant recipients: A potential target for prevention of cardiovascular complications? Transpl. Proc. 2006, 38, 2314–2316. [Google Scholar] [CrossRef]
  71. Vincenti, F.; Jensik, S.C.; Filo, R.S.; Miller, J.; Pirsch, J. A long-term comparison of tacrolimus (FK506) and cyclosporine in kidney transplantation: Evidenc for improved allograft survival at five years. Transplantation 2002, 73, 775–782. [Google Scholar] [CrossRef] [PubMed]
  72. Tory, R.; Sachs-Barrable, K.; Goshko, C.B.; Hill, J.S.; Wasan, K.M. Tacrolimus-induced elevation in plasma triglyceride concentrations after administration to renal transplant patients is partially due to a decrease in lipoprotein lipase activity and plasma concentrations. Transplantation 2009, 88, 62–68. [Google Scholar] [CrossRef] [PubMed]
  73. Ponticelli, C. The pros and the cons of mTOR inhibitors in kidney transplantation. Expert Rev. Clin. Immunol. 2014, 10, 295–305. [Google Scholar] [CrossRef] [PubMed]
  74. Kurdi, A.; Martinet, W.; de Meyer, G.R.Y. mTOR inhibition & cardiovascular diseases: Dyslipidemia and atherosclerosis. Transplantation 2017, 102, S44–S46. [Google Scholar]
  75. Andrés, V.; Castro, C.; Campistol, J.M. Potential role of proliferation signal inhibitors on atherosclerosis in renal transplant patients. Nephrol. Dial. Transpl. 2006, 21 (Suppl. 3), iii14–iii17. [Google Scholar] [CrossRef] [Green Version]
  76. Morrisett, J.D.; Abdel-Fattah, G.; Hoogeveen, R.; Mitchell, E.; Ballantyne, C.M.; Pownall, H.J.; Opekun, A.R.; Jaffe, J.S.; Oppermann, S.; Kahan, B.D. Effects of sirolimus on plasma lipids, lipoprotein levels, and fatty acid metabolism in renal transplant patients. J. Lipid Res. 2002, 43, 1170–1180. [Google Scholar] [CrossRef] [Green Version]
  77. Kobashigawa, J.A.; Kasiske, B.L. Hyperlipidemia in solid orga transplantation. Transplantation 1997, 63, 331–338. [Google Scholar] [CrossRef]
  78. Bobulescu, I.A. Renal lipid metabolism and lipotoxicity. Curr. Opin. Nephrol. Hypertens. 2010, 19, 393–402. [Google Scholar] [CrossRef] [Green Version]
  79. Izquierdo-Lahuerta, A.; Martínez-García, C.; Medina-Gómez, G. Lipotoxicity as a trigger factor of renal disease. J. Nephrol. 2016, 29, 603–610. [Google Scholar] [CrossRef]
  80. Wahl, P.; Ducasa, G.M.; Fornoni, A. Systemic and renal lipids in kidney disease development and progression. Am. J. Physiol. Ren. Physiol. 2016, 310, F433–F445. [Google Scholar] [CrossRef] [Green Version]
  81. Muntner, P.; Coresh, J.; Smith, J.C.; Eckfeldt, J.; Klag, M.J. Plasma lipids and risk of developing renal dysfunction: The atherosclerosis risk in communities study. Kidney Int. 2000, 58, 293–301. [Google Scholar] [CrossRef] [Green Version]
  82. Kang, H.M.; Ahn, S.H.; Choi, P.; Ko, Y.A.; Han, S.H.; Chinga, F.; Park, A.S.; Tao, J.; Sharma, K.; Pullman, J.; et al. Defective fatty acid oxidation in renal tubular epithelial cells has a key role in kidney fibrosis development. Nat. Med. 2015, 21, 37–46. [Google Scholar] [CrossRef] [PubMed]
  83. Carvalho, M.F.; Soares, V. Hyperlipidemia as a risk factor of renal allograft function impairment. Clin. Transpl. 2001, 15, 48–52. [Google Scholar] [CrossRef] [PubMed]
  84. Roodnat, J.I.; Mulder, P.G.; Zietse, R.; Rischen-Vos, J.; van Riemsdijk, I.C.; IJzermans, J.N.; Weimar, W. Cholesterol as an independent predictor of outcome after renal transplantation. Transplantation 2000, 69, 1704–1710. [Google Scholar] [CrossRef] [PubMed]
  85. Su, X.; Zhang, L.; Lv, J.; Wang, J.; Hou, W.; Xie, X.; Zhang, H. Effect of statins on kidney disease outcomes: A systematic review and meta-analysis. Am. J. Kidney Dis. 2016, 67, 881–892. [Google Scholar] [CrossRef] [Green Version]
  86. Geng, Q.; Ren, J.; Song, J.; Li, S.; Chen, H. Meta-analysis of the effect of statins on renal function. Am. J. Cardiol. 2014, 114, 562–570. [Google Scholar] [CrossRef]
  87. Ponticelli, C.; Arnaboldi, L.; Moroni, G.; Corsini, A. Treatment of dyslipidemia in kidney transplantation. Expert Opin. Drug Saf. 2020, 19, 257–267. [Google Scholar] [CrossRef]
  88. Brennan-Speranza, T.C.; Henneicke, H.; Gasparini, S.J.; Blankenstein, K.I.; Heinevetter, U.; Cogger, V.C.; Svistounov, D.; Zhang, Y.; Cooney, G.J.; Buttgereit, F.; et al. Osteoblasts mediate the adverse effects of glucocorticoids on fuel metabolism. J. Clin. Investig. 2012, 122, 4172–4189. [Google Scholar] [CrossRef] [Green Version]
  89. Geer, E.B.; Islam, J.; Buettner, C. Mechanisms of glucocorticoid-induced insulin resistance: Focus on adipose tissue function and lipid metabolism. Endocrinol. Metab. Clin. N. Am. 2014, 43, 75–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Ferris, H.A.; Kahn, C.R. New mechanisms of glucocorticoid-induced insulin resistance: Make no bones about it. J. Clin. Investig. 2012, 122, 3854–3857. [Google Scholar] [CrossRef] [PubMed]
  91. Ozbay, L.A.; Smidt, K.; Mortensen, D.M.; Carstens, J.; Jørgensen, K.A.; Rungby, J. Cyclosporin and tacrolimus impair insulin secretion and transcriptional regulation in INS-1E beta-cells. Br. J. Pharmacol. 2011, 162, 136–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Chakkera, H.A.; Kudva, Y.; Kaplan, B. Calcineurin Inhibitors: Pharmacologic Mechanisms Impacting Both Insulin Resistance and Insulin Secretion Leading to Glucose Dysregulation and Diabetes Mellitus. Clin. Pharmacol. Ther. 2017, 101, 114–120. [Google Scholar] [CrossRef] [PubMed]
  93. Rodriguez-Rodriguez, A.E.; Donate-Correa, J.; Rovira, J.; Cuesto, G.; Luis-Ravelo, D.; Fernandes, M.X.; Acevedo-Arozena, A.; Diekmann, F.; Acebes, A.; Torres, A.; et al. Inhibition of the mTOR pathway: A new mechanism of β cell toxicity induced by tacrolimus. Am. J. Transpl. 2019, 19, 3240–3249. [Google Scholar] [CrossRef] [PubMed]
  94. Li, Z.; Sun, F.; Zhang, Y.; Chen, H.; He, N.; Chen, H.; Song, P.; Wang, Y.; Yan, S.; Zheng, S. Tacrolimus Induces Insulin Resistance and Increases the Glucose Absorption in the Jejunum: A Potential Mechanism of the Diabetogenic Effects. PLoS ONE 2015, 10, e0143405. [Google Scholar] [CrossRef]
  95. Barlow, A.D.; Nicholson, M.L.; Herbert, T.P. Evidence for rapamycin toxicity in pancreatic β-cells and a review of the underlying molecular mechanisms. Diabetes 2013, 62, 2674–2682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Lamming, D.W.; Ye, L.; Katajisto, P.; Goncalves, M.D.; Saitoh, M.; Stevens, D.M.; Davis, J.G.; Salmon, A.B.; Richardson, A.; Ahima, R.S.; et al. Rapamycin-induced insulin resistance is mediated by mTORC2 loss and uncoupled from longevity. Science 2012, 335, 1638–1643. [Google Scholar] [CrossRef] [Green Version]
  97. Blagosklonny, M.V. Fasting and rapamycin: Diabetes versus benevolent glucose intolerance. Cell Death Dis. 2019, 10, 607. [Google Scholar] [CrossRef] [Green Version]
  98. Fioretto, P.; Mauer, M. Histopathology of diabetic nephropathy. Semin. Nephrol. 2007, 27, 195–207. [Google Scholar] [CrossRef] [Green Version]
  99. Conte, C.; Secchi, A. Post-transplantation diabetes in kidney transplant recipients: An update on management and prevention. Acta Diabetol. 2018, 55, 763–779. [Google Scholar] [CrossRef]
  100. Corremans, R.; Vervaet, B.A.; D’Haese, P.C.; Neven, E.; Verhulst, A. Metformin: A Candidate Drug for Renal Diseases. Int. J. Mol. Sci. 2018, 20, 42. [Google Scholar] [CrossRef] [Green Version]
  101. De Broe, M.E.; Kajbaf, F.; Lalau, J.D. Renoprotective Effects of Metformin. Nephron 2018, 138, 261–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Stephen, J.; Anderson-Haag, T.L.; Gustafson, S.; Snyder, J.J.; Kasiske, B.L.; Israni, A.K. Metformin use in kidney transplant recipients in the United States: An observational study. Am. J. Nephrol. 2014, 40, 546–563. [Google Scholar] [CrossRef] [PubMed]
  103. Salpeter, S.R.; Greyber, E.; Pasternak, G.A.; Salpeter, E.E. Risk of fatal and nonfatal lactic acidosis with metformin use in type 2 diabetes mellitus. Cochrane Database Syst. Rev. 2010, 2010, CD002967. [Google Scholar]
  104. Trinkley, K.E.; Anderson, H.D.; Nair, K.V.; Malone, D.C.; Saseen, J.J. Assessing the incidence of acidosis in patients receiving metformin with and without risk factors for lactic acidosis. Ther. Adv. Chronic. Dis. 2018, 9, 179–190. [Google Scholar] [CrossRef] [PubMed]
  105. Villanueva, G.; Baldwin, D. Rosiglitazone therapy of posttransplant diabetes mellitus. Transplantation 2005, 80, 1402–1405. [Google Scholar] [CrossRef]
  106. Yasmin, S.; Jayaprakash, V. Thiazolidinediones and PPAR orchestra as antidiabetic agents: From past to present. Eur. J. Med. Chem. 2017, 126, 879–893. [Google Scholar] [CrossRef]
  107. Marso, S.P.; Daniels, G.H.; Brown-Frandsen, K.; Kristensen, P.; Mann, J.F.; Nauck, M.A.; Nissen, S.E.; Pocock, S.; Poulter, N.R.; Ravn, L.S.; et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N. Engl. J. Med. 2016, 375, 311–322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Gerstein, H.C.; Colhoun, H.M.; Dagenais, G.R.; Diaz, R.; Lakshmanan, M.; Pais, P.; Probstfield, J.; Botros, F.T.; Riddle, M.C.; Rydén, L.; et al. Dulaglutide and renal outcomes in type 2 diabetes: An exploratory analysis of the REWIND randomised, placebo-controlled trial. Lancet 2019, 394, 131–138. [Google Scholar] [CrossRef]
  109. Consoli, A.; Formoso, G. Potential side effects to GLP-1 agonists: Understanding their safety and tolerability. Expert Opin. Drug Safety 2015, 14, 207–218. [Google Scholar] [CrossRef] [PubMed]
  110. Thangavelu, T.; Lyden, E.; Shivaswamy, V. A Retrospective Study of Glucagon-Like Peptide 1 Receptor Agonists for the Management of Diabetes After Transplantation. Diabetes Ther. 2020, 11, 987–994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Screen, A.J. The safety of gliptins: Updated data in 2018. Expert Opin. Drug Safety 2018, 17, 387–405. [Google Scholar]
  112. Abdelaziz, T.S.; Ali, A.Y.; Fatty, M. Efficacy and Safety of Dipeptidyl Peptidase-4 Inhibitors in Kidney Transplant Recipients with Post-transplant Diabetes Mellitus (PTDM)- a Systematic Review and Meta-Analysis. Curr. Diabetes Rev. 2020, 16, 580–585. [Google Scholar] [CrossRef] [PubMed]
  113. Perkovic, V.; Jardine, M.J.; Neal, B.; Bompoint, S.; Heerspink, H.J.L.; Charytan, D.M.; Edwards, R.; Agarwal, R.; Bakris, G.; Bull, S.; et al. Canagliflozin and Renal Outcome in Type 2 Diabetes and Nephropathy. N. Engl. J. Med. 2019, 380, 2295–2306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Anderson, S.; Cotiguala, L.; Tischer, S.; Park, J.M.; McMurry, K. Review of Newer Antidiabetic Agents for Diabetes Management in Kidney Transplant Recipients. Ann. Pharmacother. 2021, 55, 496–508. [Google Scholar] [CrossRef]
  115. Chewcharat, A.; Prasitlumkum, N.; Thongprayoon, C.; Bathini, T.; Medaura, J.; Vallabhajosyula, S.; Cheungpasitporn, W. Efficacy and Safety of SGLT-2 Inhibitors for Treatment of Diabetes Mellitus among Kidney Transplant Patients: A Systematic Review and Meta-Analysis. Med. Sci. 2020, 8, 47. [Google Scholar] [CrossRef]
  116. Feig, D.I.; Kang, D.H.; Johnson, R.J. Uric acid and cardiovascular risk. N. Engl. J. Med. 2008, 359, 1811–1821. [Google Scholar] [CrossRef]
  117. Huang, H.; Huang, B.; Li, Y.; Huang, Y.; Li, J.; Yao, H.; Wang, J. Uric acid and risk of heart failure: A systematic review and meta-analysis. Eur. J. Heart Fail 2014, 16, 15–24. [Google Scholar] [CrossRef] [PubMed]
  118. Iseki, K.; Ikemiya, Y.; Inoue, T.; Iseki, C.; Kinjo, K.; Takishita, S. Significance of hyperuricemia as a risk factor for developing ESRD in a screened cohort. Am. J. Kidney Dis. 2004, 44, 642–650. [Google Scholar] [CrossRef]
  119. Sonoda, H.; Takase, H.; Dohi, Y.; Kimura, G. Uric acid levels predict future development of chronic kidney disease. Am. J. Nephrol. 2011, 33, 352–357. [Google Scholar] [CrossRef]
  120. Zoppini, G.; Targher, G.; Chonchol, M.; Ortalda, V.; Abaterusso, C.; Pichiri, I.; Bonora, E. Serum uric acid levels and incident chronic kidney disease in patients with type 2 diabetes and preserved kidney function. Diabetes Care 2012, 35, 99–104. [Google Scholar] [CrossRef] [Green Version]
  121. Tsai, C.-W.; Lin, S.-Y.; Kuo, C.-C.; Huang, C.-C. Serum Uric Acid and Progression of Kidney Disease: A Longitudinal Analysis and Mini-Review. PLoS ONE 2017, 12, e0170393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Rodenbach, K.E.; Schneider, M.F.; Furth, S.L.; Moxey-Mims, M.M.; Mitsnefes, M.M.; Weaver, D.J.; Warady, B.A.; Schwartz, G.J. Hyperuricemia and Progression of CKD in Children and Adolescents: The Chronic Kidney Disease in Children (CKiD) Cohort Study. Am. J. Kidney Dis. 2015, 66, 984–992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Bandukwala, F.; Huang, M.; Zaltzman, J.S.; Nash, M.M.; Prasad, G.V.R. Association of uric acid with inflammation, progressive renal allograft dysfunction and post-transplant cardiovascular risk. Am. J. Cardiol. 2009, 103, 867–871. [Google Scholar] [CrossRef] [PubMed]
  124. Haririan, A.; Noguiera, J.M.; Zandi-Nejad, K.; Aiyer, R.; Hurley, H.; Cooper, M.; Weir, M.R. The independent association between serum uric acid and graft outcomes after kidney transplantation. Transplantation 2010, 89, 573–579. [Google Scholar] [CrossRef] [PubMed]
  125. Kim, D.G.; Choi, H.Y.; Kim, H.Y.; Lee, E.J.; Huh, K.H.; Kim, M.S.; Nam, C.M.; Kim, B.S.; Kim, Y.S. Association between post-transplant serum uric acid levels and kidney transplantation outcomes. PLoS ONE 2018, 13, e0209156. [Google Scholar] [CrossRef] [PubMed]
  126. Kalil, R.S.; Carpenter, M.A.; Ivanova, A.; Gravens-Mueller, L.; John, A.A.; Weir, M.R.; Pesavento, T.; Bostom, A.G.; Pfeffer, M.A.; Hunsicker, L.G. Impact of Hyperuricemia on Long-term Outcomes of Kidney Transplantation: Analysis of the FAVORIT Study. Am. J. Kidney Dis. 2017, 70, 762–769. [Google Scholar] [CrossRef]
  127. Messerli, F.H.; Frohlich, E.D.; Dreslinski, G.R.; Suarez, D.H.; Aristimuno, G.G. Serum uric acid in essential hypertension: An indicator of renal vascular involvement. Ann. Intern. Med. 1980, 93, 817–821. [Google Scholar] [CrossRef] [PubMed]
  128. Isaka, Y.; Takabatake, Y.; Takahashi, A.; Saitoh, T.; Yoshimori, T. Hyperuricemia-induced inflammasome and kidney diseases. Nephrol. Dial. Transpl. 2016, 31, 890–896. [Google Scholar] [CrossRef] [Green Version]
  129. Liu, J.; Wei, Q.; Guo, C.; Liu, Y.; Tang, C.; Dong, Z. Hypoxia, HIF, and Associated Signaling Networks in Chronic Kidney Diseases. Int J Mol Sci. 2017, 18, 950. [Google Scholar] [CrossRef]
  130. Yang, Z.; Xiaohua, W.; Lei, J.; Ruoyun, T.; Mingxia, X.; Weichun, H.; Li, F.; Ping, W.; Junwei, Y. Uric acid increases fibronectin synthesis through upregulation of lysyl oxidase expression in rat renal tubular epithelial cells. Am. J. Physiol. Renal Physiol. 2010, 299, F336–F346. [Google Scholar] [CrossRef] [Green Version]
  131. Ryu, E.-S.; Kim, M.J.; Shin, H.-S.; Jang, Y.H.; Choi, H.S.; Jo, I.; Johnson, R.J.; Kang, D.H. Uric acid-induced phenotypic transition of renal tubular cells as a novel mechanism of chronic kidney disease. Am. J. Physiol. Renal Physiol. 2013, 304, F471–F480. [Google Scholar] [CrossRef] [PubMed]
  132. Ponticelli, C.; Podestà, M.A.; Moroni, G. Hyperuricemia as a trigger of immune response in hypertension and chronic kidney disease. Kidney Int. 2020, 98, 1149–1159. [Google Scholar] [CrossRef] [PubMed]
  133. Stamp, L.K.; Day, R.O.; Yun, J. Allopurinol hypersensitivity: Investigating the cause and minimizing the risk. Nat. Rev. Rheumatol. 2016, 12, 235–242. [Google Scholar] [CrossRef] [PubMed]
  134. Kamel, B.; Graham, G.G.; Williams, K.M.; Pile, K.D.; Day, R.O. Clinical Pharmacokinetics and Pharmacodynamics of Febuxostat. Clin. Pharmacokinet. 2017, 56, 459–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Chewcharat, A.; Chang, Y.T.; Thongprayoon, C.; Crisafio, A.; Bathini, T.; Mao, M.A.; Cheungpasitporn, W. Efficacy and safety of febuxostat for treatment of asymptomatic hyperuricemia among kidney transplant patients: A meta-analysis of observational studies. Clin. Transpl. 2020, 34, e13820. [Google Scholar] [CrossRef]
  136. Choukroun, G.; Kamar, N.; Dussol, B.; Etienne, I.; Cassuto-Viguier, E.; Toupance, O. Correction of Postkidney Transplant Anemia Reduces Progression of Allograft Nephropathy. J. Am. Soc. Nephrol. 2011, 23, 360–368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Halimi, J.M.; Giraudeau, B.; Cacès, E.; Nivet, H.; Lebranchu, Y.; Tichet, J. Effects of current smoking and smoking discontinuation on renal function and proteinuria in the general population. Kidney Int. 2000, 58, 1285–1292. [Google Scholar] [CrossRef] [Green Version]
  138. Hogan, S.L.; Colindres, R.E.; Cai, J.; Coresh, J. Association of smoking with albuminuria in a cross-sectional probability sample of U.S. adults. J. Am. Soc. Nephrol. 2001, 12, 209. [Google Scholar]
  139. Samuelsson, O.; Attman, P.O. Is smoking a risk factor for progression of chronic renal failure? Kidney Int. 2000, 58, 2597. [Google Scholar] [CrossRef] [Green Version]
  140. Sung, R.S.; Althoen, M.; Howell, T.A.; Ojo, A.O.; Merion, R.M. Excess risk of renal allograft loss associated with cigarette smoking. Transplantation 2001, 71, 1752–1757. [Google Scholar] [CrossRef] [Green Version]
  141. Doyle, S.E.; Matas, A.J.; Gillingham, K.; Rosenberg, M.E. Predicting clinical outcome in the elderly renal transplant recipient. Kidney Int. 2000, 57, 2144–2150. [Google Scholar] [CrossRef] [Green Version]
  142. Kheradmand, A.; Shahbazian, H. The role of pretransplant smoking on allograft survival in kidney recipients. Urol. J. 2005, 2, 36–39. [Google Scholar] [PubMed]
  143. Nogueira, J.M.; Haririan, A.; Jacobs, S.C.; Cooper, M.; Weir, M.R. Cigarette smoking, kidney function, and mortality after live donor kidney transplant. Am. J. Kidney Dis. 2010, 55, 907–915. [Google Scholar] [CrossRef] [PubMed]
  144. Zitt, N.; Kollerits, B.; Neyer, U.; Mark, W.; Heininger, D.; Mayer, G.; Lhotta, K. Cigarette smoking and chronic allograft nephropathy. Nephrol. Dial. Transpl. 2007, 22, 3034–3039. [Google Scholar] [CrossRef] [PubMed]
  145. Suneja, M.; Khan, A.; Katz, D.A.; Kalil, R.; Nair, R. Nodular glomerulosclerosis in a kidney transplant recipient who smokes. Am. J. Kidney Dis. 2007, 50, 830–833. [Google Scholar] [CrossRef]
  146. Aker, S.; Ivens, K.; Guon, Z.; Brabensee, B.; Heering, P. Cardiovascular complications after renal transplantation. Transpl. Proc. 1998, 30, 2039–2042. [Google Scholar] [CrossRef]
  147. Orth, S.R.; Hallan, S.I. Smoking: A risk factor for progression of chronic kidney disease and for cardiovascular morbidity and mortality in renal patients—Absence of evidence or evidence of absence? Clin. J. Am. Soc. Nephrol. 2008, 3, 226–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Masiero, L.; Puoti, F.; Bellis, L.; Lombardini, L.; Totti, V.; Angelini, M.L.; Spazzoli, A.; Costa, A.N.; Cardillo, M.; Sella, G.; et al. Physical activity and renal function in the Italian kidney transplant population. Ren. Fail. 2020, 42, 1192–1204. [Google Scholar] [CrossRef] [PubMed]
  149. Kang, A.W.; Garber, C.E.; Eaton, C.B.; Risica, P.M.; Bostom, A.G. Physical Activity and Cardiovascular Risk among Kidney Transplant Patients. Med. Sci. Sports Exerc. 2019, 51, 1154–1161. [Google Scholar] [CrossRef]
  150. Mani, L.Y.; Seif, M.; Nikles, F.; Vogel, D.W.T.; Diserens, G.; Martirosian, P.; Burnier, M.; Vogt, B.; Vermathen, P. Hip Position Acutely Affects Oxygenation and Perfusion of Kidney Grafts as Measured by Functional Magnetic Resonance Imaging Methods—The Bent Knee Study. Front. Med. 2021, 8, 697055. [Google Scholar] [CrossRef]
  151. Hasse, J.M. Nutrition assessment and support of organ transplant recipients. J. Parenter. Enteral. Nutr. 2001, 25, 120–131. [Google Scholar] [CrossRef] [PubMed]
  152. Fouque, D.; Kalantar-Zadeh, K.; Kopple, J. A proposed nomenclature and diagnostic criteria for protein-energy wasting in acute and chronic kidney disease. Kidney Int. 2008, 73, 391–398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Cano, N.; Fiaccadori, E.; Tesinsky, P.; Toigo, G.; Druml, W.; Kuhlmann, M.; Mann, H.; Horl, W.H. ESPEN Guidelines on Enteral Nutrition: Adult Renal Failure. Clin. Nutr. 2006, 25, 295–310. [Google Scholar] [CrossRef] [PubMed]
  154. Ikizler, T.A.; Burrowes, J.D.; Byham-Gray, L.D.; Campbell, K.L.; Carrero, J.J.; Chan, W.; Fouque, D.; Friedman, A.N.; Ghaddar, S.; Goldstein-Fuchs, D.J.; et al. KDOQI Clinical Practice Guideline for Nutrition in CKD: 2020 Update. Am. J. Kidney Dis. 2020, 3, S1–S107. [Google Scholar] [CrossRef] [PubMed]
  155. Martínez-González, M.A.; Salas-Salvadó, J.; Estruch, R.; Corella, D.; Fitó, M.; Ros, E.; Predimed Investigators. Benefits of the Mediterranean Diet: Insights From the PREDIMED Study. Prog. Cardiovasc. Dis. 2015, 58, 50–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Martini, D. Health Benefits of Mediterranean Diet. Nutrients 2019, 11, 1802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Esposito, K.; Maiorino, M.I.; Bellastella, G.; Chiodini, P.; Panagiotakos, D.; Giugliano, D. A journey into a Mediterranean diet and type 2 diabetes: A systematic review with meta-analyses. BMJ Open. 2015, 5, e008222. [Google Scholar] [CrossRef] [Green Version]
  158. Martín-Peláez, S.; Fito, M.; Castaner, O. Mediterranean Diet Effects on Type 2 Diabetes Prevention, Disease Progression, and Related Mechanisms. A Review. Nutrients 2020, 12, 2236. [Google Scholar] [CrossRef] [PubMed]
  159. Sayón-Orea, C.; Razquin, C.; Bulló, M.; Corella, D.; Fitó, M.; Romaguera, D.; Vioque, J.; Alonso-Gómez, Á.M.; Wärnberg, J.; Martínez, J.A.; et al. Effect of a Nutritional and Behavioral Intervention on Energy-Reduced Mediterranean Diet Adherence Among Patients with Metabolic Syndrome: Interim Analysis of the PREDIMED-Plus Randomized Clinical Trial. JAMA 2019, 322, 1486–1499. [Google Scholar] [CrossRef]
  160. Mentella, M.C.; Scaldaferri, F.; Ricci, C.; Gasbarrini, A.; Miggiano, G.A.D. Cancer and Mediterranean Diet: A Review. Nutrients 2019, 11, 2059. [Google Scholar] [CrossRef] [Green Version]
  161. Gomes-Neto, A.W.; Osté, M.C.J.; Sotomayor, C.G.; van den Berg, E.; Geleijnse, J.M.; Berger, S.P.; Gans, R.O.B.; Bakker, S.J.L.; Navis, G.J. Mediterranean Style Diet and Kidney Function Loss in Kidney Transplant Recipients. Clin. J. Am. Soc. Nephrol. 2020, 15, 238–246. [Google Scholar] [CrossRef]
  162. Schmaldenst, S.; Dittrich, E.; Horl, W.H. Urinary tract infections after renal transplantation. Curr. Opin. Urol. 2002, 12, 125–130. [Google Scholar] [CrossRef]
  163. Graversen, M.E.; Dalgaard, L.S.; Jensen Fangel, S.; Jespersen, B.; Ostergaard, L.; Sogaard, O.S. Risk and outcome of pyelonephritis among renal transplant recipients. BMC Infect. Dis. 2016, 16, 264. [Google Scholar] [CrossRef] [Green Version]
  164. Ramos, E.; Drachenberg, C.B.; Papadimitriou, J.C.; Hamze, O.; Fink, J.C.; Klassen, D.K.; Drachenberg, R.C.; Wiland, A.; Wali, R.; Cangro, C.B.; et al. Clinical course of polyoma virus nephropathy in 67 renal transplant patients. J. Am. Soc. Nephrol. 2002, 13, 2145–2151. [Google Scholar] [CrossRef] [Green Version]
  165. Vats, A. BK virus-associated transplant nephropathy: Need for increased awareness in children. Pediatr. Transpl. 2004, 8, 421–425. [Google Scholar] [CrossRef]
  166. Hirsch, H.H.; Knowles, W.; Dickermann, M. Prospective studies of polyomavirus type BK replication and nephropathy in renal transplant recipients. N. Engl. J. Med. 2002, 347, 527–530. [Google Scholar] [CrossRef]
  167. Nickeleit, V.; Singh, H.K.; Mihatsch, M.J. Polyomavirus nephropathy: Morphology, pathophysiology, and clinical management. Curr. Opin. Nephrol. Hypertens. 2003, 12, 599–606. [Google Scholar] [CrossRef] [PubMed]
  168. Kiberd, B.A. Screening to prevent polyoma virus nephropathy: A medical decision analysis. Am. J. Transpl. 2005, 10, 2410–2416. [Google Scholar] [CrossRef] [PubMed]
  169. Randhawa, P.S.; Vats, A.; Zygmunt, D.; Swalsky, P.; Scantlebury, V.; Shapiro, R.; Finkelstein, S. Quantitation of viral DNA in renal allograft tissue from patients with BK virus nephropathy. Transplantation 2002, 74, 485–488. [Google Scholar] [CrossRef] [Green Version]
  170. Johnston, O.; Jaswal, D.; Gill, J.S.; Doucette, S.; Fergusson, D.A.; Knoll, G.A. Treatment of polyomavirus infection in kidney transplant recipients: A systematic review. Transplantation 2010, 89, 1057–1070. [Google Scholar] [CrossRef] [Green Version]
  171. Berger, S.P.; Sommerer, C.; Witzke, O.; Tedesco, H.; Chadban, S.; Mulgaonkar, S.; Qazi, Y.; de Fijter, J.W.; Oppenheimer, F.; Cruzado, J.M.; et al. Two-year outcomes in de novo renal transplant recipients receiving everolimus-facilitated calcineurin inhibitor reduction regimen from the TRANSFORM study. Am. J. Transpl. 2019, 19, 3018–3034. [Google Scholar] [CrossRef] [PubMed]
  172. Available online: https://www.anzdata.org.au/report/anzdata-43rd-annual-report-2020-anzdata/ (accessed on 1 April 2022).
Figure 1. CNI renal toxicity. (a) CNI arteriolopathy; a preglomerular arteriole showing mucinoid thickening of the arteriolar wall. (b) CNI arteriolopathy with severe nodular hyalinosis of the wall. (c) Striped interstitial fibrosis, tubular dilatation and atrophy. (d) Diffuse interstitial fibrosis, glomerular ischemia and sclerosis.
Figure 1. CNI renal toxicity. (a) CNI arteriolopathy; a preglomerular arteriole showing mucinoid thickening of the arteriolar wall. (b) CNI arteriolopathy with severe nodular hyalinosis of the wall. (c) Striped interstitial fibrosis, tubular dilatation and atrophy. (d) Diffuse interstitial fibrosis, glomerular ischemia and sclerosis.
Jpm 12 01271 g001
Figure 2. Main causes of post-transplant hypertension.
Figure 2. Main causes of post-transplant hypertension.
Jpm 12 01271 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ponticelli, C.; Citterio, F. Non-Immunologic Causes of Late Death-Censored Kidney Graft Failure: A Personalized Approach. J. Pers. Med. 2022, 12, 1271. https://doi.org/10.3390/jpm12081271

AMA Style

Ponticelli C, Citterio F. Non-Immunologic Causes of Late Death-Censored Kidney Graft Failure: A Personalized Approach. Journal of Personalized Medicine. 2022; 12(8):1271. https://doi.org/10.3390/jpm12081271

Chicago/Turabian Style

Ponticelli, Claudio, and Franco Citterio. 2022. "Non-Immunologic Causes of Late Death-Censored Kidney Graft Failure: A Personalized Approach" Journal of Personalized Medicine 12, no. 8: 1271. https://doi.org/10.3390/jpm12081271

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop