Next Article in Journal
Discovery of TBX20 as a Novel Gene Underlying Atrial Fibrillation
Previous Article in Journal
From Chaos to Opportunity: Decoding Cancer Heterogeneity for Enhanced Treatment Strategies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Hyperglycemia and Hyperlipidemia with Kidney or Liver Transplantation: A Review

by
John A. D’Elia
and
Larry A. Weinrauch
*
Kidney and Hypertension Section, E P Joslin Research Laboratory, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
*
Author to whom correspondence should be addressed.
Biology 2023, 12(9), 1185; https://doi.org/10.3390/biology12091185
Submission received: 27 July 2023 / Accepted: 22 August 2023 / Published: 29 August 2023

Abstract

:

Simple Summary

Post-allograft transplant antirejection regimens (glucocorticoids, azathioprine, mycophenolate, calcineurin inhibitors and mTOR inhibitors) may trigger or aggravate hyperglycemia or hyperlipidemia. Post-transplant medication management must balance immune suppression and glucose and lipid control because the existence of glucose or lipid imbalance is associated with shorter times of useful allograft function. We review the underlying mechanism of relationships between glycemia/lipidemia control, transplant rejection and graft aging.

Abstract

Although solid organ transplantation in persons with diabetes mellitus is often associated with hyperglycemia, the risk of hyperlipidemia in all organ transplant recipients is often underestimated. The diagnosis of diabetes often predates transplantation; however, in a moderate percentage of allograft recipients, perioperative hyperglycemia occurs triggered by antirejection regimens. Post-transplant prescription of glucocorticoids, calcineurin inhibitors and mTOR inhibitors are associated with increased lipid concentrations. The existence of diabetes mellitus prior to or following a liver transplant is associated with shorter times of useful allograft function. A cycle involving Smad, TGF beta, m-TOR and toll-like receptors has been identified in the contribution of rejection and aging of allografts. Glucocorticoids (prednisone) and calcineurin inhibitors (cyclosporine and tacrolimus) induce hyperglycemia associated with insulin resistance. Azathioprine, mycophenolate and prednisone are associated with lipogenesis. mTOR inhibitors (rapamycin) are used to decrease doses of atherogenic agents used for immunosuppression. Post-transplant medication management must balance immune suppression and glucose and lipid control. Concerns regarding rejection often override those relative to systemic and organ vascular aging and survival. This review focuses attention on the underlying mechanism of relationships between glycemia/lipidemia control, transplant rejection and graft aging.

1. Introduction

Despite diabetes mellitus being the most prevalent underlying condition requiring transplantation, all solid organ recipients are subject to hyperlipidemic vascular complications. Underlying mechanisms explaining vascular risk associated with immunosuppressive antirejection medication have not been widely disseminated.
Type 2 diabetes mellitus, predominantly a state of insulin resistance with increased lipogenesis, is initially associated with hyperinsulinemia that may decrease with time. We now recognize subtypes of type 2 diabetes that may be characterized by remission: gestational remitting with delivery, obesity-related remitting with bariatric surgery and steroid or immunosuppression related remitting with therapy cessation. In some patients, the appearance of hyperglycemia exposes a trait that may lead to the well-known complications of diabetes (nephropathy, neuropathy, retinopathy and early vascular disease). However, in others, prolonged follow-up does not result in these complications. The prognostic impact of these subtypes within the transplant population is unknown.
In the 50 years of genetically non-related kidney transplantation, there was a gradual acceptance of recipients with diabetes mellitus, followed by awareness that hyperglycemia and infection were important side effects of immunosuppression [1]. The US Renal Data System report for 2000–2018 included 70,000 persons with diabetes mellitus who had received a kidney transplant [2]. The survivals for 50,000 persons with type 2 diabetes were 94% at 1 year, 77% at 5 years, 48% at 10 years and 25% at 15 years [2].
Individuals with type 1 diabetes mellitus were younger, demonstrating higher survivals at these time intervals (96%, 85%, 65% and 46%). Among over 10,000 persons with native kidneys, diabetes and hypertension, modulation of medication dosages for control of hyperglycemia was noted to be related to the estimated glomerular filtration rate [3]. Modulations of immunosuppressive regimens were also observed in the FAVORIT study involving 1500 kidney transplant recipients with diabetes mellitus morbidity [4]. Demonstration that infection and malignancy were the most frequent causes of post-transplant mortality among diabetic transplant recipients prompted the quest to eliminate causes of hyperglycemia [5]. Whether acquired diabetes mellitus is found before or after kidney transplantation, the mechanism of vascular complications relates to extracellular glycation of protein and lipid components of cell structures with cross-linking that alters function [6,7]. Several immunosuppressive agents (prednisone, calcineurin inhibitors and mTor antagonists) contribute to post-transplant hyperglycemia. Recently appreciated intracellular alterations involving protein kinases (especially series of protein kinase C) have become the focus of attention.

2. Mammalian Target of Rapamycin in Diabetes Mellitus: Nephropathy and Kidney Transplantation

Mammalian target of rapamycin (mTOR) is a complex of protein kinase signal molecules that promote kidney disease. Located in the glomerular podocyte and mesangial cell, mTOR is involved in the generation of diabetic nephropathy (Table 1). The mTOR complex located in the renal tubular epithelial cell is involved in the generation of polycystic kidney disease [8,9]. The mTOR inhibitor sirolimus (rapamycin) was discovered 50 years ago. Sirolimus has immunosuppressive effects as well as anti-tumor and anti-fungal effects [10]. Two viruses best known for complicating the course of kidney allograft recipients are cytomegalovirus (CMV) and BK virus. Both viruses can injure allograft function. Kidney allograft host cells provide activated mTOR for CMV and BKV replication. Several studies suggest that anti-mTOR immunosuppressive agents have antiviral benefits as opposed to other immunosuppressive medications [11,12,13]. Following receipt of renal allografts, the prevalence of squamous cell skin cancer is lower with mTOR inhibitors than with other immunosuppressive agents [10]. Because combinations of immunosuppressive agents may be required, calcineurin inhibitors (such as cyclosporine), which can be nephrotoxic and diabetogenic at high doses, may be combined with mTOR Inhibitors. Some investigations recommend a dose of cyclosporine low enough to avoid nephrotoxicity to the allograft along with a dose of rapamycin low enough to avoid lipotoxicity to the heart [14,15,16].

3. Hyperglycemia Associated with Medications for Transplant Immunosuppression

Three excellent articles introduce the topic of medications for immunosuppression with the timeline of post-transplantation hyperglycemia [17,18,19]. To the best of our knowledge, azathioprine and mycophenolate do not generally cause hyperglycemia or hyperlipidemia. Transplant allograft recipients treated with azathioprine may have increased concentrations of triglycerides (Table 1) [20]. Mycophenolate mofetil treatment reduces the risk of plaque formation in the atherosclerosis-prone rabbit on a high-cholesterol diet [21].
The medication most often associated with hyperglycemia or the concept of “new onset diabetes after transplantation” in the years after kidney transplantation is prednisone [22]. Mechanisms for insulin resistance and allograft injury with glucocorticoids include steroids that make the liver less sensitive to insulin, so it carries on releasing glucose even if the pancreas is releasing insulin [23]; steroids that cause increased appetite, fluid retention and weight gain; and increased gluconeogenesis—the conversion of muscle protein amino acids into glucose stored in the liver as glycogen. This side effect is not as potent as lipogenesis in the problem of insulin resistance (Table 2). In addition, there is increased lipogenesis, the conversion of muscle protein amino acids into fatty acids stored in visceral adipose tissue and the liver as triglycerides. Osteocalcin, secreted by osteoblasts, functions to promote bone strength. Another function of osteocalcin is the suppression of lipogenesis in the liver and visceral adipose tissue. Therefore, the use of glucocorticoids, which inhibit the action of osteocalcin, also results in lipogenesis with resultant insulin resistance [24,25]. Finally, toxic hyperglycemia in human [26] and experimental animal [27] renal tubular epithelial cells involve caspase enzymes that generate hydrogen peroxide.
As a result of the mechanisms listed in Table 2, tacrolimus (and cyclosporine, both calcineurin inhibitors) decreases insulin secretion by the suppression of expression of genes for beta cell function, resulting in a decreased level of insulin mRNA [28] and inhibition of the activity of glucokinase, thereby decreasing Beta cell response to increases in glucose concentration [29] and reducing the number of pancreatic Beta cells by inducing apoptosis of Beta cells [30].
Rapamycin and other mTOR inhibitors eliminate the insulin resistance associated with protein kinase-related hypertriglyceridemia [31]. This mechanism has been studied extensively in the streptozotocin (STZ) diabetic rat (type 1 diabetes model). In this model, kidney glomerular mesangial cells exposed to increased levels of glucose were associated with increased activity of Protein Kinase C (PKC) beta isoform [32]. Activated PKC beta isoforms were associated with increased expression of Transforming Growth Factor (TGF) Beta. TGF Beta is known to be associated with increased production of fibronectin and type 4 collagen (Table 3), which leads to mesangial expansion and basement membrane thickening in glomerular cells in diabetes mellitus. Pericytes attached to capillaries in the retina are potentially susceptible to hyperglycemia associated with advanced glycated end-products, which cross-link pathologically [33]. Additional studies found pathological changes in the vascular layer of the retina [34]. The progression of microvascular changes (loss of perivascular support cells and capillary aneurysms) has been documented in both the STZ diabetic rat and the transgenic mouse. Inhibitors of PKC isoform beta have been shown to prevent pathological findings in the renal glomerulus and ophthalmic retina [35,36,37].
The Banff Human Organ Transplantation study identified the peritubular portion of the capillary as the target for antibody attack in chronic active transplant rejection of kidney allografts [38,39]. The process is referred to as capillaritis (Table 4). The well-known cardiac complication of hyperglycemia, cardiomyopathy, considered to be the result of the cross-linking of advanced glycated end-products, causes stiffness in relaxation and contraction [40]. A gene factor for remodeling in diabetic cardiomyopathy, known as Suppressor of mothers against decapentaplegic (Smad-3), operates in cooperation with TGF-Beta [41]. Weakness in retinal capillaries leading to aneurysms may be due to the inflexibility of retinal pericytes. Thus, capillaries in kidney allografts now shown to be the target of antibodies in chronic active rejection may also be the subject of weakening through cross-linking of advanced glycated end-products (Table 4).
Investigators have described a cascade: Smad → TGF Beta → mTOR → mesangial collagen fibrosis inhibited by rapamycin [42]. This rapamycin effect could be over-ridden by hypoxia-inducible factor (HIF) 1 alpha at normal oxygen levels.
Further investigation into the role of the important member of mTOR, the protein kinase PKC, was centered upon early diabetic nephropathy (albuminuria) in the STZ diabetic rat. At this early stage, innate immunity (toll-like receptors) was activated along with cytokines of inflammation (interleukin 17). Inhibition of Protein Kinase C by rapamycin was associated with the blockade of progression of nephropathy in this experimental model of Type 1 diabetes mellitus [35]. There is an mTOR tuberin pathway. Tuberin inhibits cell cycle progression and, therefore, has a negative impact on the life cycle of renal tubular cells. Hyperglycemia contributes to the apoptosis of renal tubular epithelial cells under the influence of tuberin and is blocked by insulin [36]. In the mTOR/tuberin pathway, cell size regulation likely occurs by activation/inhibition of tuberin vs. inhibition/activation of mTOR [36]. Thus, the insulin Akt cascade both activates and downgrades tuberin with opposing effects on MTOR, thereby regulating cell size.

4. Lipogenesis before/after Transplantation

4.1. Kidney Allografts

Pericardiac and perirenal fat are predictors of atherosclerotic pathology affecting cardiovascular outcomes and renal function. When perirenal fat volume was studied, live donor renal grafts with higher fat volume resulted in lower estimated GFR than those with lesser perirenal fat volume [43].
Cholesterol is essential for the structure of the glomerular podocyte diaphragm, which functions to select molecules for excretion versus those to be retained. Excess levels of ceramide, a sphingolipid, can be destructive to the ability to discriminate solutes critical for excretion from those appropriate for retention [44]. Increased concentrations of oxidized low-density lipoproteins are toxic to podocytes of glomeruli in membranous and diabetic nephropathy, both of which may recur allograft during follow-up [45]. Excess levels of triglyceride may inhibit tubular resorption of electrolytes and amino acids [46]. A deficiency of peroxisome proliferator-activated receptor (PPAR) gamma has been found to have increased expression of nuclear factor kappa beta associated with inflammation/fibrosis [47]. Among experimental animals exposed to thiazolidinediones, synthetic PPAR gamma agonists, kidney function was preserved [47].
It stands to reason that some of these pre-transplantation mechanisms will be operating after transplantation, depending upon the choice of immunosuppression [48]. The provision of pancreas allograft in addition to kidney allograft appears to produce better cardiovascular outcomes than kidney transplant alone among diabetic recipients [48,49,50,51,52]. Differences in endothelial-dependent dilation of the brachial artery and intimal thickness of the carotid artery [48,49] may contribute to the differences observed in long-term follow-up. Hyperlipidemia experimental models are available for studies of azathioprine and mycophenolate [20,21] as noted in Table 1.

4.2. Liver Allografts

Although liver insufficiency does not commonly occur after a kidney transplant, renal insufficiency may occur after a liver transplant. Among twelve liver allograft recipients studied before and after transplant, an observed fall in the glomerular filtration rate (GFR (27%) was not due to dehydration or heart failure as evidenced by increases in both somatic blood flow (RBF) and oxygen utilization following transplantation [53].
Follow-up of recipients of liver allografts indicates significantly better results for those without a diagnosis of diabetes mellitus compared to those with diabetes mellitus [54]. Pretransplant adult-onset diabetes occurring as liver function decreases to a state of fibrosis known as cirrhosis and has been termed “hepatogenous” diabetes [55]. Glucose tolerance testing pre- and post-transplant has demonstrated a new type 2 diabetes post-transplant [56]. Hepatic lipotoxicity is associated with cholesterol, ceramides, sphingolipids and triglycerides. These lipid molecules may be encapsulated into droplets as intracellular organelles that may cause mitochondrial injury and stress the endoplasmic reticulum [57,58,59].
Since some of the mechanisms of lipogenesis are associated with excess nutrition or elevated glucocorticoid dose, there is a chance for recurrence of the process after transplantation. Excessive caloric intake or toxic ingestion results in steatosis/fibrosis of the allograft. There is no evidence for allograft-related lipogenesis [60].
Glucocorticoids contribute to lipogenesis by the conversion of skeletal muscle amino acids through acetate to glycogen via glycogen synthase. Similarly, glucocorticoids organize the conversion of skeletal muscle amino acid through acetate to palmitate with the stimulation of fatty acid synthase [61,62,63]. Nonalcoholic fatty liver disease is an important cause of liver failure. Since this metabolic condition is associated with insulin resistance, it will recur in liver transplant allografts, especially if glucocorticoid immunosuppressive doses are high [64,65]. The calcineurin inhibitor, cyclosporin, was associated with elevated levels of serum cholesterol, which were reduced when another calcineurin inhibitor, tacrolimus, was introduced into the transplantation immunosuppressive program [66].

5. Management of Post-Transplant Hyperglycemia

Minimal to moderate hyperglycemia following solid-organ transplantation may not require treatment and may not ordinarily be called post-transplant diabetes mellitus [67,68]. A key report indicated that the post-kidney or post-liver-transplantation new onset of diabetes mellitus may be as high as 20% in each instance [29]. Three publications involving the term post-transplantation diabetes mellitus pinpoint calcineurin inhibitors (tacrolimus and cyclosporine) as fundamental since glucocorticoid doses would have been significantly diminished [19,69,70,71], establishing the definition of post-transplantation diabetes mellitus as continued hyperglycemia beyond 45 days (6+ weeks) of treatment, emphasizing the key mechanism of toxicity of cyclosporine to be the removal of the cell surface glucose transporter. A Lewis rat model was described, quantifying the recurrence of coronary artery atherosclerosis in heart transplant allografts [71]. In this study, both groups received cyclosporine and a high-fructose diet to induce post-heart transplant hyperlipidemia. However, only the group rendered insulin-dependent by the use of streptozotocin developed severe coronary artery atherosclerosis in short-term follow-up [71].
When kidney allograft function falls below an estimated glomerular filtration of 60 mL/minute (Table 5), there is a need to reduce doses of certain sulfonylurea medications (glyburide), dipeptidyl dipeptidase receptor 4 inhibitors (sitagliptin), biguanides (metformin) and insulin since these medications are renally excreted. The sulfonyl urea glipizide is cleared by liver function. Since sodium/glucose co-transporter 2 inhibitor (canagliflozin, empagliflozin and dapagliflozin) efficiency is reduced at this lower end of the estimated glomerular filtration rate, prescription is not recommended. On the other hand, there is active consideration for the use of glucagon-like peptide 1 receptor agonists (exenatide, liraglutide, dulaglutide) in the setting of calcineurin inhibition or tacrolimus-related hyperglycemia [72,73].
Since the estimated glomerular filtration rate (eGFR) may change rapidly with unrelated kidney allografts, doses of antihyperglycemic medications may require rapid change. Insulin is metabolized and excreted by the kidney, and levels rise with deceasing allograft function. C-Peptide, also renally excreted, is a more reliable measure of circulating insulin that is removed in part with hepatic passage through the liver. The glucose/C-Peptide ratio is useful in decisions of doses of medications for glucose control, especially when eGFR is changing [74]. The normal level for glucose/C-Peptide is 55 ± 25 mg/dL/ng/mL (range of 30–80). A high glucose/C-Peptide concentration (>120) is consistent with the need to initiate insulin injections in addition to the other classes being used. A low glucose/C-Peptide ratio (<20) is consistent with hypoglycemia unless all anti-hyperglycemic medication doses are lowered.
In rare instances, all doses of medications for glucose control are discontinued as eGFR is falling [75], leading to the return to dialysis dependency for the kidney transplant recipient.

6. Management of Post-Transplant Hyperlipidemia

Post-transplant prescription of glucocorticoids (prednisone and dexamethasone), calcineurin inhibitors (cyclosporin and tacrolimus) and mTOR inhibitors (sirolimus and everolimus) is associated with increased lipid concentrations. Glucocorticoids increase triglycerides through the inhibition of lipoprotein lipase, calcineurin inhibitors also inhibitors lipoprotein lipase [76]. Lower levels of post-transplantation lipids have been identified with belatacept. The mechanism is associated with a pathway involving the co-stimulation of antigen-bearing B lymphocytes and antibody-producing T lymphocytes [76].
In addition to the well-known adverse cardiovascular outcomes of long-term hyperlipidemia and their reduction with lipid-lowering agents, there is now significant research linking elevated lipids to allograft rejection and vasculopathy [76,77,78,79,80,81,82,83]. Mechanisms for allograft vasculopathy include the activation of smooth muscle endothelium [77,78,79]. The presence of hyperlipidemia is associated with excess vascular plaque formation. The reduction in serum lipid levels among transplant recipients has been demonstrated to decrease accelerated allograft vasculopathy [84]. Pharmacologic lipid reduction achieved by the use of HMG CoA reductase inhibitors may reduce antibody-mediated rejection by the inhibition of T cell proliferation and T cell interaction with antigen-bearing B lymphocytes [85]. Apolipoprotein E (Apo E) mediates the uptake of cholesterol through an ldl receptor pathway. Deletion of apo E results in a significant increase in plasma cholesterol due to failure to clear the lipoprotein. Apo E is made in macrophages, not T or B cells. In the mouse model, the deletion of apo E is associated with impaired uptake of cholesterol in the liver and elevated plasma cholesterol when compared to wild-type mice. When both types are exposed to high-fat diets, the apoE-deficient mice have higher cholesterol, and rejection of cardiac allograft occurs more rapidly. Exposure of the wild type to a high-fat diet with a slower rise in plasma cholesterol is associated with similar but slower evidence for rejection [86]. Cardiac allografts are rejected equally in wild-type vs. apo E-deficient mice, confirming the human observation of increased rejection risk associated with hyperlipidemia.
Hyperlipidemia following solid organ transplant is a consequence of the interaction of immunosuppressive agents, genetic predisposition and diet/obesity [87]. Each group of immunosuppressive agents that have been used in solid organ recipients (corticosteroids, azathioprine, mycophenolate mofetil, calcineurin inhibitors and mTor antagonists) either alone or in combination has been demonstrated to potentiate dyslipidemia and adversely affect long-term allograft outcomes.
Despite significant animal model and human research linking adverse cardiovascular outcomes to hyperlipidemia, there are no allograft-recipient-specific guidelines for the control of hyperlipidemia.
Generally, kidney and liver transplant candidates are chosen because their risk of cardiovascular events is low. For this reason, trials of preventive therapies often do not have sufficient cardiovascular events to produce meaningful statistical differences in benefits [5]. For patients in whom the allograft is cardiac, however, the need for lowering of lipid and plaque burden is more evident. Based upon the current literature and our observation, we feel that primary prevention through lipid-lowering therapy is indicated when prolonged survival should be anticipated and hyperlipidemia, hyperglycemia or frank diabetes is noted. There is also support for lipid-lowering therapy in all heart transplant recipients or when allograft function is threatened. Given the multiple medications that must be taken by organ recipients, careful attention must be paid to possible drug–drug interactions and the fact that research is relatively limited to HMG CoA reductase inhibitors (statins). For this reason, we would favor outcome studies that focus on the best serum lipid levels for transplant recipients for the preservation of allograft function and avoidance of cardiovascular events. With respect to the choice of lipid-lowering agents, one must be aware of drug–drug interactions with various immunomodulators (especially cyclosporine) and the relative lack of information demonstrating comparative benefits of newer agents when compared to the most studied HMG Co-A reductase inhibitors (statins). With respect to newer agents such as ezetimibe, PCSK9 inhibitors and small interfering RNA-based therapeutics, there are few outcome trials in organ transplant recipients to support their widespread use.

7. Conclusions

The presence of diabetes or hyperlipidemia of either long duration or accompanying the requirement for organ replacement therapy is associated with elevated risk to allograft function [86,87]. Glycemic control is more difficult with the use of prednisone and tacrolimus [88]. The control of hyperlipidemia is more difficult with the use of azathioprine and high-dose sirolimus. Elevated lipid levels contribute to insulin resistance. Elevated glucose levels lead to advanced glycated end-products, which may cross-link, causing stiffness, i.e., resistance to movement. The restriction of relaxation and contraction (cardiomyopathy) is the best-known result of stiffness associated with advanced glycated end-products. This process may involve pericytes around capillaries as well. Pericyte stiffness would leave the capillary at risk for the formation of micro-aneurysms, which are seen in diabetic retinopathy.
Increased risk for kidney allograft rejection is partially due to increased activity of the inflammation cascade. Activation of the pathway known as mammalian target of rapamycin occurs in persons with diabetes mellitus. This leads to increased inflammation, which contributes to allograft rejection. The Banff Transplantation Study has identified the capillary pericyte as the most sensitive site for antibody attack in chronic, active allograft rejection. We can conclude that a diagnosis of diabetes mellitus, before or after the initiation of immunosuppression, contributes to the risk of loss of allograft function, with the capillary pericyte as a major focus of pathogenesis. A cascade combining several factors in kidney fibrosis included sMAD → TGF Beta → mTOR → mesangial collagen. Rapamycin was protective by blocking the process of fibrosis, but the rapamycin effect could be over-ridden by the over-expression of HIF (Figure 1).
Beyond the scope of this review are gestational diabetes [89], obesity treated with bariatric surgery [90,91] and the treatment of renal allograft recipients with mTOR inhibition who received the SARS-CoV-2 mRNA BNT 162 bz (Pfizer BioNTech) vaccine. Their antibody response was greater than that of matched controls who did not receive mTOR inhibitors [92]. Also, beyond the scope of this review is consensus on the definition of remission of type 2 diabetes mellitus [93,94], which may be useful as transplant recipients are treated with a variety of immunosuppressive agents. We also have little information to clarify the relation between immunosuppression and observed combinations of diabetic nephropathy and membranous glomerulopathy sometimes found in renal allografts [95,96,97].

Author Contributions

J.A.D. and L.A.W. contributed early in all phases of this manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

Not applicable.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Tony Monaco was a pioneer in kidney transplantation of persons with diabetes mellitus. When the Harvard 5th Surgical Service moved from Boston City Hospital to the New England Deaconess Hospital in 1973, Tony’s office was in proximity to the Joslin Diabetes Center. We had the pleasure and honor to collaborate with someone firmly committed to the application of new research to new clinical practice. Tony was still active this year despite extraordinary physical challenges, a model not unlike that of Elliot Joslin who worked a half-day the day he passed away at age 92 years. We dedicate this paper to the memory of a fearless surgeon willing to consider transplanting our patients with diabetes mellitus despite their multiple comorbidities.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bach, M.C.; Adler, J.L.; Breman, J.; P’eng, F.K.; Sahyoun, A.; Schlesinger, R.M.; Madras, P.; Monaco, A.P. Influence of rejection therapy on fungal and nocardial infections in renal-transplant recipients. Lancet 1973, 1, 180–184. [Google Scholar] [CrossRef]
  2. Harding, J.L.; Pavkov, M.; Wang, Z.; Benoit, S.; Burros, N.R.; Imperatore, G.; Albright, A.L.; Patzer, R. Long-term mortality among kidney transplant recipients with and without diabetes: A nationwide cohort study in the USA. BMJ Open Diabetes Res. Care 2021, 9, e001962. [Google Scholar] [CrossRef]
  3. Weinrauch, L.A.; Segal, A.R.; Bayliss, G.; Liu, J.; Wisniewski, E.; D’Elia, J.A. Changes in treatment of hyperglycemia in a hypertensive type 2 diabetes population as renal function declines. Clin. Kidney J. 2017, 10, 661–665. [Google Scholar] [CrossRef]
  4. Weinrauch, L.A.; D’Elia, J.A.; Weir, M.R.; Bunnapradist, S.; Finn, P.; Liu, J.; Claggett, B.; Monaco, A.P. Does diabetes impact therapeutic immune modulation therapy decisions for kidney transplant recipients? Data from the Folic Acid for Vascular Outcome Reduction in Transplant (FAVORIT) trial. Int. J. Nephrol. Renov. Dis. 2017, 10, 233–242. [Google Scholar] [CrossRef] [PubMed]
  5. Weinrauch, L.A.; D’Elia, J.A.; Weir, M.R.; Bunnapradist, S.; Finn, P.V.; Liu, J.; Claggett, B.; Monaco, A.P. Infection and malignancy outweigh cardiovascular mortality in kidney transplant recipients: Post hoc analysis of the FAVORIT trial. Am. J. Med. 2018, 131, 165–172. [Google Scholar] [CrossRef]
  6. Yamagishi, S.; Takeuchi, M.; Inagaki, Y.; Imaizumi, T. Role of advanced glycation end products (AGEs) and their receptor (RAGE) in the pathogenesis of diabetic microangiopathy. Int. J. Clin. Pharm. Res. 2003, 23, 129–134. [Google Scholar]
  7. Jud, P.; Sourij, H. Therapeutic options to reduce advanced glycated end products in patients with diabetes mellitus: A review. Diabetes Res. Clin. Prac. 2019, 148, 54–63. [Google Scholar] [CrossRef] [PubMed]
  8. Fantus, D.; Rogers, N.M.; Grahammer, F.; Huber, T.B.; Thomson, A.W. Roles of mTOR complexes in the kidney: Implication for kidney disease and transplantation. Nat. Rev. Nephrol. 2016, 12, 587–609. [Google Scholar] [CrossRef]
  9. Viana, S.D.; Reis, F.; Alves, R. Therapeutic use of m-TOR inhibitors in renal diseases: Advances, drawbacks, and challenges. Oxid. Med. Cell Longev. 2018, 2018, 3693625. [Google Scholar] [CrossRef]
  10. Wolf, S.; Hoffmann Vs Habicht, A.; Kauke, T.; Bucher, J.; Schoenberg, M.; Werner, J.; Guba, M.; Andrassy, J. Effect of small mTOR-is on malignancy and survival following renal transplantation: A systematic review and meta-analysis of randomized trials with a minimum follow up of 24 months. PLoS ONE 2018, 13, e0194975. [Google Scholar] [CrossRef]
  11. Zeng, J.; Zhong, G.; Feng, X.; Li, I.; Fang, S.; Fan, Y.; Song, T.; Huang, Z.; Wang, X.; Lin, T. Conversion from calcineurin inhibitors to mammalian target of rapamycin kidney transplant recipients: A systematic and meta-analysis of randomized controlled trials. Front. Immunol. 2021, 12, 663602. [Google Scholar] [CrossRef] [PubMed]
  12. Cucchiari, D.; Rios, J.; Molina-Andujar, A.; Montagud-Marrahi, E.; Revuelta, J.; Ventura-Aguiar, P.; Pineiro, G.P.; De Souza-Amorin, E.; Esforzado, N.; Cofan, F.; et al. Combination of calcineurin-mTor inhibitor in kidney transplantation: A propensity score analysis. J. Nephrol. 2020, 33, 601–610. [Google Scholar] [CrossRef] [PubMed]
  13. Verges, B.; Cariou, B. mTor inhibitors and diabetes. Diabetes Res. Clin. Prac. 2015, 110, 101–108. [Google Scholar] [CrossRef] [PubMed]
  14. Schena, F.P.; Gesualdo, L. Pathogenetic mechanisms of diabetic nephropathy. J. Am. Soc. Nephrol. 2005, 16 (Suppl. S1), s30–s33. [Google Scholar] [CrossRef] [PubMed]
  15. Mori, H.; Inoki, K.; Masutani, K.; Wakabayashi, Y.; Komai, K.; Nakagawa, R.; Guan, K.L.; Yoshimura, A. The mTOR pathway is highly activated I diabetic nephropathy and rapamycin has a strong therapeutic potential. Biochem. Biophys. Res. Commun. 2009, 384, 471–475. [Google Scholar] [CrossRef]
  16. Flechner, S.M. mTor inhibition and clinical transplantation. Transplantation 2018, 102, 517–518. [Google Scholar] [CrossRef]
  17. Li, J.X.; Cummins, C.L. Fresh insights into glucocorticoid induced diabetes mellitus and new therapeutic directions, Nat. Rev. Endocrinol. 2022, 18, 540–555. [Google Scholar]
  18. Kuo, T.; McQueen, A.; Chen, T.C.; Wang, J.C. Regulation of glucose homeostasis by glucocorticoids. Adv. Exp. Med. Biol. 2015, 872, 99–126. [Google Scholar]
  19. Midtvedt, K.; Hjelmesaeth, J.; Hartmann, A.; Lund, K.; Paulsen, D.; Egeland, T.; Jenssen, T. Insulin resistance after renal transplantation: The effect of steroid dose reduction and withdrawal. J. Am. Soc. Nephrol. 2004, 15, 3233–3239. [Google Scholar] [CrossRef]
  20. Akman, B.; Uyar, M.; Afsar, B.; Sezer, S.; Ozdemir, F.N.; Haberal, M. Lipid profile during azathioprine or mycophenolate mofetil combination with cyclosporine and steroids. Transplant. Proc. 2007, 39, 135–137. [Google Scholar] [CrossRef]
  21. Romero, F.; Rodriguez-Iturbe, B.; Pong, H.; Parra, G.; Rincon, J.; Gonzalez, I. Mycophenolate mofetil treatment reduces cholesterol-induced atherosclerosis in the rabbit. Atherosclerosis 2000, 152, 127–133. [Google Scholar] [CrossRef] [PubMed]
  22. Munshi, V.N.; Saghafian, S.; Cook, C.B.; Werner, K.T.; Chakkera, H.A. Comparison of post- transplantation diabetes mellitus and risk factors between kidney and liver transplantation patients. PLoS ONE 2020, 15, e0226873. [Google Scholar] [CrossRef] [PubMed]
  23. 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] [PubMed]
  24. Brennan-Speranza Henneicke, H.; Gasparini, S.J.; Blankenstein, K.I.; Heinevetter, U.; Cogger, V.C.; Svistounov, D.; Zhang, V.; Cooney, G.J.; Buttgereit, F.; Dunstan, C.R.; et al. Osteoblasts mediate the adverse effect of glucocorticoids on fuel metabolism. J. Clin. Invest. 2012, 112, 4172–4189. [Google Scholar] [CrossRef]
  25. Ferris, H.A.; Kahn, C.R. New mechanisms of glucocorticoid-induced insulin resistance: Make no bones about it. Clin. Invest. 2012, 112, 3854–3857. [Google Scholar] [CrossRef]
  26. Verzola, D.; Bertolotto, M.B.; Villaggio, B.; Ottonello, L.; Dallegri, F.; Salvatore, F.; Berruti, V.; Gandolfo, M.T.; Garibotto, G.; Deferrari, G. Oxidative stress mediates apoptotic changes induced by hyperglycemia in human tubular kidney cells. J. Am. Soc. Nephrol. 2004, 15, 585–587. [Google Scholar] [CrossRef]
  27. Allen, D.A.; Harwood, S.; Varagunam, M.; Raftery, M.J.; Yaqoob, M.M. High glucose-induced oxidative stress causes apoptosis in proximal tubular epithelial cells and is mediated by multiple caspases. FASEB J. 2003, 17, 908–910. [Google Scholar] [CrossRef]
  28. De Graav, G.N.; van der Zwan, M.; Baan, C.C.; Janssen, J.A.M.J.L.; Hesselink, D.A. Improved glucose tolerance in a kidney transplant recipient with type 2 diabetes mellitus from tacrolimus to belatacept: A case report and review of mechanisms. Transplant. Direct 2018, 4, e350. [Google Scholar] [CrossRef]
  29. Kukla, A.; Hill, J.; Merzkani, A.; Lorenz, E.C.; Park, E.D.; D’Costa, M.; Kudva, Y.C.; Stengel, M.D.; Shah, P. The use of GLP 1R agonists for treatment of type 2 diabetes mellitus in kidney transplant recipients. Transplant. Direct 2020, 6, e524. [Google Scholar] [CrossRef]
  30. Pavlakis, M.; Goldfarb-Rumyantzev, A.S. Diabetes after transplantation and sirolimus: What’s the connection? J. Am. Soc. Nephrol. 2008, 19, 1255–1256. [Google Scholar] [CrossRef]
  31. Tremblay, F.; Marette, A. Amino acid and insulin signaling via the mTOR/p70 56 kinase pathway: A negative feedback mechanism leading to insulin resistance in skeletal muscle cells. J. Biol. Chem. 2001, 27, 38052–38060. [Google Scholar] [CrossRef]
  32. Koya, D.; Jirousek, M.R.; Lin, Y.W.; Ishii, H.; Kuboki, K.; King, G.L. Characterization of Protein Kinase C beta isoform activation on the gene expression of transforming growth factor beta, extracellular matrix components, and prostanoids in the glomeruli of diabetic rats. J. Clin. Invest. 1997, 100, 115–126. [Google Scholar] [CrossRef] [PubMed]
  33. Koya, D.; King, G.L. Protein Kinase C activation in the development of diabetes mellitus complications. Diabetes 1998, 47, 859–866. [Google Scholar] [CrossRef]
  34. Lang, G.E. Pharmacological treatment of diabetic retinopathy. Ophthalmologica 2007, 221, 112–117. [Google Scholar] [CrossRef] [PubMed]
  35. Yu, R.; Bo, H.; Vilani, V.; Spencer, P.H.; Fu, P. Inhibitory effect of rapamycin on toll-like receptor 4 and interleukin 17 in the early—Stage of diabetic nephropathy. Kidney Blood Press. Res. 2016, 41, 55–69. [Google Scholar] [CrossRef] [PubMed]
  36. Velagapudi, M.; Bhandari, B.S.; Abboud-Werner, S.; Simone, S.; Abboud, H.E.; Habib, S.L. The tuberin/mTOR pathway promotes apoptosis of tubular epithelial cells in diabetes. J. Am. Soc. Nephrol. 2011, 22, 262–273. [Google Scholar] [CrossRef]
  37. Rosner, M.; Freilinger, A.; Hengstschlager, M. Akt regulates nuclear/cytoplasmic localization of tuberin. Oncogene 2007, 26, 521–531. [Google Scholar] [CrossRef]
  38. Rosales, I.A.; Mahowald, G.K.; Tomaszewski, K.; Hotta, K.; Iwahara, N.; Otsuka, T.; Tsuji, T.; Takada, Y.; Acheampong, E.; Araujo-Medina, M.; et al. Peritubular capillaritis in chronic active antibody-mediated rejection. J. Am. Soc. Nephrol. 2022, 33, 2306–2319. [Google Scholar] [CrossRef]
  39. Bissonnette, M.L.Z.; Riazy, M.; Cunningham, A.M.; Gill, J.S. A step toward understanding the story behind the pictures: Molecular diagnostics and the Banff Classification of renal allograft pathology. J. Am. Soc. Nephrol. 2022, 33, 2131–2132. [Google Scholar] [CrossRef]
  40. D’Elia, J.A.; Bayliss, G.; Weinrauch, L.A. The diabetic cardio renal nexus. Int. J. Mol. Sci. 2022, 23, 7351. [Google Scholar] [CrossRef]
  41. Dobaczewski, M.; Chen, W.; Frangogiannis, N.G. Transforming growth factor-Beta (TGF)-B signaling in cardiac remodeling. J. Mol. Cell. Cardiol. 2011, 51, 600–606. [Google Scholar] [CrossRef]
  42. Rozen-Zvi, B.; Hayashida, T.; Hubchak, C.B.; Hanna, C.; Platanias, L.C.; Schnaper, H.W. TGF-B/Smad3 activates mammalian target of rapamycin comples-1 o promote collagen produced by increasing HIF 1a expression. Am. J. Physiol. Renal Physiol. 2013, 305, F485–F494. [Google Scholar] [CrossRef]
  43. Tastemur, S.; Olcucuoglu, E.; Karaaslan, M. High perirenal fat volume affects negatively renal function in living renal transplantation. Transplant. Proc. 2022, 54, 1768–1772. [Google Scholar] [CrossRef]
  44. Bobulescu, I. Renal lipid metabolism and lipotoxicity. Curr. Opin. Nephrol. Hypertens. 2010, 19, 393–402. [Google Scholar] [CrossRef] [PubMed]
  45. Feldcamp, T.; Kribben, A.; Roeser, N.; Senter, R.A.; Weinberg, J.M. Accumulation of fatty acids causes the sustained energetic deficit in kidney proximal tubules after hypoxia-re-oxygenation. Am. J. Physiol. Renal Physiol. 2006, 290, F465–F477. [Google Scholar] [CrossRef] [PubMed]
  46. Feldcamp, T.; Weinberg, J.; Horbelt, M.; Von Kropff, C.; Witzke, O.; Nurnberger, J.; Kribben, A. Evidence for involvement of non-esterified fatty acid-induced protonophoric uncoupling during mitochondrial dysfunction caused by hypoxia and re-oxygenation. Nephrol. Dial. Transplant. 2009, 24, 43–51. [Google Scholar] [CrossRef] [PubMed]
  47. Sun, Y.; Ge, X.; Li, X.; He, J.; Wei, X.; Du, J.; Sun, J.; Li, X.; Xun, Z.; Liu, W.; et al. High fat diet promotes renal injury by inducing oxidative stress and mitochondrial dysfunction. Cell Death Dis. 2020, 11, 914. [Google Scholar] [CrossRef]
  48. Biesenbach, G.; Margreiter, R.; Konigsrainer, A.; Bosmuller, C.; Janko, O.; Brucke, P.; Gross, C.; Zazgornik, J. Comparison of progression of macrovascular diseases after kidney or pancreas and kidney transplantation in patients with end-stage renal disease. Diabetologia 2000, 43, 231–234. [Google Scholar] [CrossRef]
  49. Biesenbach, G.; Konigsrainer, A.; Gross, C.; Margreiter, R. Progression of macrovascular disease is reduced in type 1 diabetic patients after more than five years successful combined pancreas-kidney transplantation in comparison to kidney transplantation alone. Transpl. Int. 2005, 18, 1054–1060. [Google Scholar] [CrossRef]
  50. Lombardo, C.; Perrone, V.G.; Amorese, G.; Vistoli, F.; Baronti, W.; Marchetti, P.; Boggi, V. Update on Pancreatic Transplantation in the Management of Diabetes in Endotext; Feingold, K.R., Anawalt, B., Blackman, M.R., Boyce, A., Chrousos, G., Corpas, E., de Herder, W.W., Dhatariya, K., Dungan, K., Hofland, J., et al., Eds.; MDText.com, Inc.: South Dartmouth, MA, USA, 2000. [Google Scholar]
  51. Fiorina, P.; LaRocca, E.; Venturini, M.; Minicucci, F.; Fermo, I.; Paroni, R.; D’Angelo, A.; Sblendido, M.; DiCarlo, V.; Cris-tallo, M.; et al. Effects of kidney-pancreas transplantation on atherosclerotic risk factors and endothelial function in patients with uremia and type 1 diabetes mellitus. Diabetes 2001, 50, 496–501. [Google Scholar] [CrossRef]
  52. Larsen, J.L.; Ratanasuwan, T.; Burkman, T.; Lynch, T.; Erickson, J.; Colling, C.; Lane, J.; Mack-Shipman, L.; Lyden, E.; Lo-seke, M.; et al. Carotid intima media thickness is decreased after pancreas transplantation. Transplantation 2002, 73, 936–940. [Google Scholar] [CrossRef]
  53. Larssen, J.S.; Bragadottir, G.; Redfors, B.; Ricksten, S.-E. Renal function and oxygenation are impaired early after liver transplantation despite hyperdynamic systemic circulation. Crit. Care 2017, 21, 87. [Google Scholar] [CrossRef] [PubMed]
  54. Li, P.; Fan, H.; He, O. Pretransplant diabetes mellitus predicts worse outcome of liver transplantation: Evidence from meta-analysis. J. Endocrinol. Invest. 2018, 41, 211–221. [Google Scholar] [CrossRef] [PubMed]
  55. Orsi, E.; Grancini, V.; Menini, S.; Aghemo, A.; Pugliese, G. Hepatogenous diabetes: Is it time to separate it from type 2 diabetes? Liver Int. 2017, 37, 950–962. [Google Scholar] [CrossRef]
  56. Kim, M.G.; Cho, W.C. Differential diagnosis of diabetes caused by liver cirrhosis and other type 2 diabetes mellitus. Korean J. Hepatol. 2006, 12, 524–529. [Google Scholar] [PubMed]
  57. Liangpunsakul, S.; Chalasani, N. Lipid mediators of liver injury in nonalcoholic fatty liver disease. Am. J. Physiol. Gastroenterol. Liver Physiol. 2019, 316, 675–681.52. [Google Scholar] [CrossRef]
  58. Mordier, S.; Iynedjian, P.B. Activation of mammalian target of rapamycin complex 1 and insulin resistance induced by palmitate in hepatocytes. Biochem. Biophys. Res. Commun. 2007, 362, 206–211. [Google Scholar] [CrossRef]
  59. Pierantonelli, I.; Svegliati-Baroni, G. Non-alcoholic fatty liver disease: Basic pathogenetic mechanisms in the progression from NAFLD to NASH. Transplantation 2019, 103, e1–e13. [Google Scholar] [CrossRef]
  60. Farrell, G.C.; Haczeyni, F.; Chitturi, S. Pathogenesis of NASH: How metabolic complications of over nutrition favors lipo-toxicity and proinflammatory fatty liver disease. Adv. Exp. Med. Biol. 2018, 1001, 19–44. [Google Scholar]
  61. Minehira, K.; Novel-Chate, V.; Schwarz, J.M.; Gillet, M.; Darioli, R.; Chiolero, R.; Tappy, L. Hepatic de novo lipogenesis after liver transplantation. J. Parenter. Enteral Nutr. 2001, 25, 229–235. [Google Scholar] [CrossRef]
  62. Stegall, M.D.; Everson, G.T.; Schroter, G.; Karrer, F.; Bilir, B.; Sternberg, T.; Shrestha, R.; Wachs, M.; Kam, I. Prednisone withdrawal late after liver transplantation reduces diabetes, hypertension, and hypercholesterolemia without causing graft loss. Hepatology 1997, 25, 173–177. [Google Scholar] [CrossRef]
  63. Wang, Y.; Voy, B.J.; Urs, S.; Kim, S.; Soltani–Bejnood, M.; Quigley, M.; Heo, Y.-R.; Standridge, M.; Andersen, B.; Dhar, M.; et al. The human fatty acid synthase gene and de novo lipogenesis are coordinately regulated in human adipose tissue. J. Nutr. 2004, 134, 1032–1038. [Google Scholar] [CrossRef]
  64. Berdanier, C.D. Role of glucocorticoids in the regulation of lipogenesis. FASEB J. 1989, 3, 2179–2183. [Google Scholar] [CrossRef] [PubMed]
  65. Jimenez-Perez, M.; Gonzalez-Grande, R.; Guzman, E.O.; Trillo, V.A.; Rodrigo-Lopez, J.M. Metabolic complications in liver transplantation recipients. World J. Gastroenterol. 2016, 22, 6416–6423. [Google Scholar] [CrossRef] [PubMed]
  66. Roy, A.; Kneteman, N.; Lilly, L.; Marotta, P.; Peltekian, K.; Scudamore, C.; Tchervenkov, J. Tacrolimus as intervention in the treatment of hyperlipidemia after liver transplantation. Transplantation 2006, 82, 494–500. [Google Scholar] [CrossRef] [PubMed]
  67. Ozbay, L.A.; Smidt, K.; Mortensen, D.M.; Carstens, J.; Jorgensen, J.A.; Rungby, J. Cyclosporin and tacrolimus impair insulin secretion and transcriptional regulation in INS-1Ebeta-cells. Br. J. Pharmacol. 2011, 162, 136–146. [Google Scholar] [CrossRef]
  68. Nordheim, E.; Birkeland, K.I.; Asberg, A.; Hartmann, A.; Horneland, R.; Jenssen, T. Preserved insulin secretion and kidney function in recipients with functional pancreas grafts 1 year after transplantation: A single-center prospective observation study. Eur. J. Endocrinol. 2018, 179, 251–259. [Google Scholar] [CrossRef]
  69. Blagosklonny, M.V. Fasting and rapamycin: Diabetes versus benevolent glucose intolerance. Cell Death Dis. 2019, 10, 607. [Google Scholar] [CrossRef]
  70. Iqbal, A.; Zhou, K.; Kashyap, S.R.; Lansang, M.B. Early renal transplant hyperglycemia. J. Clin. Endocrinol. Metab. 2022, 107, 549–562. [Google Scholar] [CrossRef]
  71. Hoang, K.; Chen, I.; Reaven, G.; Zhang, L.; Ross, H.; Billingham, M.; Valentine, H. Diabetes and dyslipidemia: A new model for transplant coronary artery disease. Circulation 1998, 97, 2160–2168. [Google Scholar] [CrossRef]
  72. Radu, R.G.; Fujimoto, S.; Mukai, E.; Takehiro, M.; Shimono, D.; Nabe, K.; Shimodahira, M.; Kominato, R.; Aramaki, Y.; Nishi, Y.; et al. Tacrolimus suppresses glucose-induced insulin from pancreatic islets by reducing glucokinase activity. Am. J. Physiol. Endocrinol. Metab. 2005, 288, E365–E371. [Google Scholar] [CrossRef]
  73. Tong, L.; Li, W.; Huang, Y.; Zhou, F.; Zhou, Y.; Zhan, L.; Li, J.; Song, Z.; Yu, M.; Zhou, C.; et al. Tacrolimus inhibits insulin release and promotes apoptosis of Mon6 cells through the inhibition of the P13k/Akt/mTOR pathway. Mol. Med. Rep. 2021, 24, 658. [Google Scholar] [CrossRef]
  74. D’Elia, J.A.; Mulla, C.; Weinrauch, L.A. Variations in Glucose/C-Peptide in persons with diabetes mellitus associated with renal function. Diabetes Res. Clin. Prac. 2019, 150, 1–7. [Google Scholar] [CrossRef] [PubMed]
  75. D’Elia, J.A.; Kaldany, A.; Miller, D.G.; Rolla, A.; Weinrauch, L.A. Elimination for requirement of exogenous insulin therapy in diabetic renal failure. Ren. Fail. 1982, 6, 75–82. [Google Scholar] [CrossRef]
  76. Chmielnicka, K.; Heleniak, Z.; Debska-Slizien, A. Dyslipidemia in renal transplant recipients. Transplantology 2022, 3, 188–199. [Google Scholar] [CrossRef]
  77. Rahmani, M.; Cruz, R.P.; Granville, D.J.; McManus, B.M. Allograft Vasculopathy Versus Atherosclerosis. Circ. Res. 2006, 99, 801–815. [Google Scholar] [CrossRef] [PubMed]
  78. Kobashigawa, J.A. Statins and cardiac allograft vasculopathy after heart transplantation. Semin. Vasc. Med. 2004, 4, 401–406. [Google Scholar] [CrossRef]
  79. Valantine, H.J. Cardiac allograft vasculopathy after heart transplantation: Risk factors and management. Heart Lung Transpl. 2004, 23 (Suppl. 5), S187–S193. [Google Scholar] [CrossRef]
  80. Bagley, J.; Williams, L.; Hyde, M.; Birriel, C.R.; Iacomini, J. Hyperlipidemia and Allograft Rejection. Curr. Transplant. Rep. 2019, 6, 90–98. [Google Scholar] [CrossRef]
  81. Opazo-Rios, L.; Mas, S.; Marin-Royo, G.; Mezzano, S.; Gomez-Guerrero, C.; Moreno, J.A.; Egido, J. Lipotoxicity and Diabetic Nephropathy: Novel mechanistic insights and therapeutic opportunities. Int. J. Mol. Sci. 2020, 21, 2632. [Google Scholar] [CrossRef]
  82. Palmer, S.C.; Navaneethan, S.D.; Craig, J.C.; Perkovic, V.; Johnson, D.W.; Nigwekar, S.U.; Hegbrant, J.; Strippoli, G.F. HMG CoA reductase inhibitors (statins) for kidney transplant recipients. Cochrane Database Syst. Rev. 2014, 2014, CD005019. [Google Scholar] [CrossRef] [PubMed]
  83. Warden, B.A.; Duell, P.B. Management of dyslipidemia in adult solid organ transplant recipients. J. Clin. Lipidol. 2019, 13, 231–245. [Google Scholar] [CrossRef]
  84. Cardarelli, F.; Saidman, S.; Theruvath, T.; Tolkoff-Rubin, N.; Cosimi, A.B.; Pascual, M. The problem of late allograft loss in kidney transplantation Minerva. Urol. Nefrol. 2003, 55, 1–11. [Google Scholar]
  85. Agarwal, A.; Prasad, G.V. Post-transplant dyslipidemia: Mechanisms, diagnosis and management. World J. Transplant. 2016, 6, 125–134. [Google Scholar] [CrossRef]
  86. Chowdhury, T.A. Post-transplant diabetes mellitus. Clin. Med. 2019, 19, 392–395. [Google Scholar] [CrossRef] [PubMed]
  87. Escasany, E.; Izquierdo-Lahuerta, A.; Medina-Gomez, G. Underlying mechanisms of renal lipotoxicity in obesity. Nephron 2019, 143, 28–32. [Google Scholar] [CrossRef]
  88. Hecking, M.; Sharif, A.; Eller, K.; Jenssen, T. Management of post-transplant diabetes: Immunosuppression, early prevention, and antidiabetics. Transpl. Int. 2021, 34, 27–48. [Google Scholar] [CrossRef] [PubMed]
  89. Crowther, C.A.; Samuel, D.; McCowan, L.M.E.; Edlin, R.; Tran, T.; McKinlay, C.J. for the GEMS Trial Group. Lower versus higher glycemic criteria for diagnosis of gestational diabetes. N. Engl. J. Med. 2022, 387, 587–598. [Google Scholar] [CrossRef]
  90. Singh, A.K.; Singh, R.; Kota, K. Bariatric surgery and diabetes remission: Who would have thought it? Indian. J. Diabetes Metab. 2015, 19, 563–576. [Google Scholar] [CrossRef]
  91. Chumacova-Orin, M.; Vanetta, C.; Moris, D.P.; Guerron, A.D. Diabetes mellitus remission after bariatric surgery. World J. Diabetes 2021, 12, 1093–1158. [Google Scholar] [CrossRef]
  92. Netti, G.S.; Infante, B.; Troise, D.; Mercuri, S.; Panico, M.; Spadaccino, F.; Catalano, V.; Gigante, M.; Simone, S.; Pontrelli, P.; et al. mTOR inhibitors improve both humeral and cellular responses to SARS-CoV-2 mRNA BNT 16b2 vaccine in kidney transplantation recipients. Am. J. Transplant. 2022, 22, 1475–1482. [Google Scholar] [CrossRef] [PubMed]
  93. Buse, J.B.; Caprio, S.; Cefalu, W.T.; Ceriello, A.; Del Prato, S.; Inzucchi, S.E.; McLaughlin, S.; Phillips GL 2nd Robertson, R.P.; Rubino, F.; Kahn, R.; et al. How do we define cure of diabetes? Diabetes Care. 2009, 32, 2133–2135. [Google Scholar] [CrossRef] [PubMed]
  94. Riddle, M.C.; Cefalu, W.T.; Evans, P.H.; Gerstein, H.C.; Nauck, M.A.; Oh, W.K.; Rothberg, A.E.; le Roux, C.W.; Rubino, F.; Schauer, P.; et al. Consensus report: Definition and interpretation of remission in type 2 diabetes. Diabetologia. 2021, 64, 2359–2366. [Google Scholar] [CrossRef] [PubMed]
  95. Truong, L.; Gelfand, J.; D’Agati, V.; Tomaszewski, J.; Appel, G.; Hardy, M.; Pirani, C.L. De novo membranous glomerulopathy in renal allografts: A review of ten cases and review of the literature. Am. J. Kidney Dis. 1989, 14, 131–144. [Google Scholar] [CrossRef]
  96. Markowitz, G. Membranous glomerulopathy: Emphasis on secondary forms and disease variants. Adv. Anat. Pathol. 2001, 8, 119–125.63. [Google Scholar] [CrossRef]
  97. Bhadauria, D.; Chellappan, A.; Kaul, A.; Etta, P.; Badri, V.; Sharma RKPrasad, N.; Gupta, A.; Jain, M. Idiopathic membranous glomerulopathy in patients with diabetes mellitus: A diagnostic and therapeutic quandary. Clin. Kidney J. 2018, 11, 46–56. [Google Scholar] [CrossRef]
Figure 1. Mechanisms of allograft rejection and aging in diabetes mellitus. * inhibited by rapamycin. AGE (Advanced Glycated End-Products). sMAD (Suppressor of Mothers against Decapentaplegic Transcription Factor Family). TGF Beta (Transforming Growth Factor Beta). mTOR (mammalian Target Of Rapamycin).
Figure 1. Mechanisms of allograft rejection and aging in diabetes mellitus. * inhibited by rapamycin. AGE (Advanced Glycated End-Products). sMAD (Suppressor of Mothers against Decapentaplegic Transcription Factor Family). TGF Beta (Transforming Growth Factor Beta). mTOR (mammalian Target Of Rapamycin).
Biology 12 01185 g001
Table 1. Metabolic Effects of Post-Organ-Transplant Immunosuppression.
Table 1. Metabolic Effects of Post-Organ-Transplant Immunosuppression.
DrugDysmetabolic EffectObservation
PrednisoneIncreased lipogenesis and gluconeogenesisRat liver study (rat model)
AzathioprineIncreased triglyceridesTransplant patients treated with azathioprine
Mycophenolate mofetilDecreased atheroma sizeAtheroma prone rabbit on atherogenic diet
Calcineurin inhibitors (cyclosporin, tacrolimus)Decreased insulin secretionHumans post-organ-transplant
mTOR * inhibitors (sirolimus, temsirolimus, everolimus)Increased insulin resistanceHepatocytes (rat), muscle cell cultures
* mTOR (mammalian target of Rapamycin).
Table 2. Pathogenesis of Complications of Diabetes Mellitus Associated with Protein Kinases.
Table 2. Pathogenesis of Complications of Diabetes Mellitus Associated with Protein Kinases.
  • Protein Kinase C: Increased levels associated with TGF Beta
    • Increased fibronectin, type 4 collagen with complications of diabetes mellitus.
      • Retina of eye: capillary aneurisms with bleeding.
      • Glomerulus of kidney: fibrosis of basement membranes, mesangial cells.
    • Increased innate immunity with rejection of kidney allografts.
      • Toll-like receptors.
      • Cytokines (interleukin 17).
    • Inhibition of Na+/K+ ATPase
      • Limits control of hypokalemia with hyper-aldosterone and hyperkalemia with ACE inhibitor.
  • mTOR *: increased levels for recipients of kidney transplant allografts.
    • Decreased insulin secretion: reversed by rapamycin.
    • Decreased production of antibodies after COVID vaccine: reversed by rapamycin.
* mTOR (mammalian target of Rapamycin).
Table 3. Contribution of mTOR * to Pathology: Protection with Rapamycin.
Table 3. Contribution of mTOR * to Pathology: Protection with Rapamycin.
  • Kidney
    • Glomerulus
      • Podocyte dysfunction.
      • Mesangial expansion.
      • Basement membrane thickening.
      • Diabetic nephropathy.
    • Tubule
      • Expression of familial polycystic disorder.
      • Injury with BK and CM ** viruses.
      • Post-transplantation injury with cyclosporine.
  • Skin
    • Post-transplantation squamous cell cancer.
* mammalian target of rapamycin. ** BK and cytomegalovirus.
Table 4. Pathogenesis of Vascular Complications for recipients of Kidney Transplants: Pre vs. Post-Transplant Diabetes Mellitus.
Table 4. Pathogenesis of Vascular Complications for recipients of Kidney Transplants: Pre vs. Post-Transplant Diabetes Mellitus.
Pathologic Anatomy
Mechanism
AGE *sMAD **TGF-Beta ***mTOR ****
Pericyte/FibrosisHyperglycemia++--
-Fibronectin++--
Collagen 4++--
Artery/Atheroma
Hyperlipidemia--++
Toll-like receptors--++
Interleukin 17--++
Cardiac Muscle Hypertrophy
Hypertension----
Fibrosis++--
* AGE Advanced Glycated End-Products. ** sMAD Suppressor of Mothers against Decapentaplegic Transcription Factor Family. *** TGF Transforming Growth Factor Beta. **** mTOR mammalian target of Rapamycin.
Table 5. Medications to Control Hyperglycemia: Pre- or post-Transplantation.
Table 5. Medications to Control Hyperglycemia: Pre- or post-Transplantation.
  • GFR > 60 mL/minute: usual doses of
    • Insulin.
    • Metformin.
    • Sulfonyl urea glyburide (glipizide).
    • Dipeptidyl peptidase receptor inhibitor (sitagliptin).
    • Glucagon-like receptor agonist (liraglutide, dulaglutide).
    • Sodium/glucose transport inhibitor 2 (canagliflozin, empagliflozin, dapagliflozin).
  • GFR 30–60 mL/minute: Reduced doses of
    • Insulin.
    • Metformin.
    • Glyburide but not glipizide.
    • Sitagliptin.
    • Liraglutide and Dulaglutide.
    • Frequently not used not used: canagliflozin, empagliflozin and dapagliflozin.
  • GFR < 30 mL/minute: Restricted doses of
    • Insulin.
    • Metformin.
    • Glyburide but not glipizide.
    • Sitagliptin.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

D’Elia, J.A.; Weinrauch, L.A. Hyperglycemia and Hyperlipidemia with Kidney or Liver Transplantation: A Review. Biology 2023, 12, 1185. https://doi.org/10.3390/biology12091185

AMA Style

D’Elia JA, Weinrauch LA. Hyperglycemia and Hyperlipidemia with Kidney or Liver Transplantation: A Review. Biology. 2023; 12(9):1185. https://doi.org/10.3390/biology12091185

Chicago/Turabian Style

D’Elia, John A., and Larry A. Weinrauch. 2023. "Hyperglycemia and Hyperlipidemia with Kidney or Liver Transplantation: A Review" Biology 12, no. 9: 1185. https://doi.org/10.3390/biology12091185

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