Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2339 2023-07-05 15:42:58 |
2 only format change Meta information modification 2339 2023-07-06 05:52:27 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Qu, L.; Jiao, B. The Impact of Immunometabolic Dysregulation in Kidney Disease. Encyclopedia. Available online: https://encyclopedia.pub/entry/46467 (accessed on 12 August 2024).
Qu L, Jiao B. The Impact of Immunometabolic Dysregulation in Kidney Disease. Encyclopedia. Available at: https://encyclopedia.pub/entry/46467. Accessed August 12, 2024.
Qu, Lili, Baihai Jiao. "The Impact of Immunometabolic Dysregulation in Kidney Disease" Encyclopedia, https://encyclopedia.pub/entry/46467 (accessed August 12, 2024).
Qu, L., & Jiao, B. (2023, July 05). The Impact of Immunometabolic Dysregulation in Kidney Disease. In Encyclopedia. https://encyclopedia.pub/entry/46467
Qu, Lili and Baihai Jiao. "The Impact of Immunometabolic Dysregulation in Kidney Disease." Encyclopedia. Web. 05 July, 2023.
The Impact of Immunometabolic Dysregulation in Kidney Disease
Edit

Kidney disease encompasses a diverse group of disorders that vary in their underlying pathophysiology, clinical presentation, and outcomes. These disorders include acute kidney injury (AKI), chronic kidney disease (CKD), glomerulonephritis, nephrotic syndrome, polycystic kidney disease, diabetic kidney disease, and many others. Despite their distinct etiologies, these disorders share a common feature of immune system dysregulation and metabolic disturbances. The immune system and metabolic pathways are intimately connected and interact to modulate the pathogenesis of kidney diseases. The dysregulation of immune responses in kidney diseases includes a complex interplay between various immune cell types, including resident and infiltrating immune cells, cytokines, chemokines, and complement factors. These immune factors can trigger and perpetuate kidney inflammation, causing renal tissue injury and progressive fibrosis.

immune metabolic inflammation kidney disease

1. Introduction

Kidney disease is a significant health problem worldwide, affecting an estimated 10% of the global population [1]. The most common forms of kidney disease include chronic kidney disease (CKD) and acute kidney injury (AKI) [2]. However, kidney disease encompasses a diverse group of disorders that vary in their underlying pathophysiology, clinical presentation, and outcomes. These disorders include tubulointerstitial, glomerulonephritis, nephrotic syndrome, polycystic kidney disease, diabetic kidney disease, vascular disease, vasculitis, and congenital kidney disease, among others.
Immunometabolism refers to the interplay between immune and metabolic pathways, which are tightly regulated in normal physiological conditions [3][4][5][6]. In pathological conditions, such as kidney disease, this delicate balance is disrupted, leading to immunometabolic dysregulation. Immunometabolic dysregulation involves various cell types, such as T cells, B cells, macrophages, and dendritic cells, as well as cytokines, chemokines, and metabolic processes, such as oxidative stress, mitochondrial dysfunction, and inflammation [4][7].

2. The Impact of Immunometabolic Dysregulation in Kidney Disease

2.1. Acute Kidney Injury (AKI)

Acute kidney injury (AKI) is a complex condition characterized by a rapid loss of renal function [8][9]. Immunometabolic dysregulation has been shown to play an important role in the pathogenesis of AKI [10][11]. This involves an imbalance between pro- and anti-inflammatory cytokines, leading to the activation of innate immune cells and subsequent tissue damage.
Several genes and pathways have been linked to immunometabolic dysregulation in AKI. One of the key pathways involved in the development of AKI is the hypoxia-inducible factor 1-alpha (HIF-1α) pathway [12][13]. Under hypoxic conditions, HIF-1α is stabilized and activates the transcription of genes involved in glycolysis, angiogenesis, and inflammation [14]. Studies have shown that HIF-1α plays a critical role in the development of AKI by promoting glycolysis in immune cells and contributing to the production of pro-inflammatory cytokines [15][16][17]. In addition, HIF-1α can also upregulate glucose transporter 1 (GLUT1), which facilitates glucose uptake in immune cells, and its upregulation has been linked to the development of AKI [17][18]. Moreover, recent studies have suggested that epigenetic modifications, such as DNA methylation and histone modifications, can contribute to the dysregulation of HIF-1α in AKI pathogenesis [19][20][21]. Another important gene involved in immunometabolic dysregulation in AKI is the gene encoding for inducible nitric oxide synthase (iNOS). iNOS is an enzyme that produces nitric oxide (NO), which is a potent regulator of immune cell function [22]. Dysregulation of iNOS has been implicated in the pathogenesis of AKI, with studies showing that iNOS-mediated NO production can contribute to tissue damage in the kidney [23][24][25].
In addition to HIF-1α and iNOS, toll-like receptors (TLRs) are involved in the recognition of pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), and their dysregulation has been linked to the development of AKI [26][27]. TLRs can activate nuclear factor kappa B (NF-κB), a transcription factor that regulates the expression of genes involved in inflammation and immune cell activation, and its dysregulation has been shown to contribute to the development of AKI [28][29]. The NLRP3 inflammasome, a multiprotein complex involved in the processing and secretion of pro-inflammatory cytokines, has also been implicated in the development of AKI. Studies have shown that NLRP3 inflammasome activation can contribute to the development of AKI by promoting the secretion of pro-inflammatory cytokines [30][31][32].

2.2. Chronic Kidney Disease (CKD)

Chronic kidney disease (CKD) is a progressive condition characterized by the gradual loss of kidney function over time. Dysregulation of immune cells and metabolism contribute to the accumulation of toxic metabolites, oxidative stress, and fibrosis, which are key contributors to the progression of CKD [33]. One of the key pathways involved in the development of CKD is dysregulated glucose metabolism in immune cells [34][35]. Studies have shown that this dysregulation can lead to the activation of pro-inflammatory pathways, oxidative stress, and endothelial dysfunction, all of which can contribute to the development of CKD [36]. GLUT1 and HIF-1α are two genes that have been implicated in the dysregulation of glucose metabolism in immune cells in the context of CKD [37][38]. Another important pathway involved in CKD is the activation of the NLRP3 inflammasome and subsequent cytokine production. Increased NLRP3 expression has been observed in patients with CKD, and inhibition of the NLRP3 inflammasome has been shown to ameliorate kidney damage in animal models of CKD [39]. Additionally, dysregulated lipid metabolism has been linked to the progression of CKD. Studies have shown that increased levels of FFAs can contribute to the development of CKD by activating inflammatory pathways and inducing oxidative stress [40][41]. In addition to the above-mentioned pathways, other genes involved in immune cell dysregulation in CKD include TLRs, NF-κB, and the renin–angiotensin–aldosterone system (RAAS). TLRs are involved in the recognition of PAMPs and DAMPs, and their dysregulation has been linked to the development of CKD [42][43][44]. NF-κB activation in CKD can be triggered by a variety of stimuli, including oxidative stress, hypoxia, and proinflammatory cytokines, such as TNF-α and IL-1β [45][46]. Furthermore, NF-κB activation is tightly linked to NLRP3 inflammasome activation in CKD. Activation of the NLRP3 inflammasome triggers the activation of NF-κB, which, in turn, leads to the production of more proinflammatory cytokines, creating a positive feedback loop that perpetuates the inflammatory response [47][48]. The RAAS is a hormone system that regulates blood pressure and fluid balance in the body, and its dysregulation has been linked to the development of CKD through its effects on renal hemodynamics and inflammation [49][50][51].

2.2.1. Lupus Nephritis

Lupus nephritis is a type of kidney inflammation that occurs as a result of systemic lupus erythematosus (SLE), an autoimmune disease [52]. Immunometabolic dysregulation is one of the key mechanisms underlying the pathogenesis of lupus nephritis [53]. Dysregulated metabolism in immune cells can contribute to the production of autoantibodies and the activation of inflammatory cells, leading to glomerular damage and renal dysfunction [54][55].
Several genes and pathways have been implicated in the dysregulated metabolism in immune cells in the context of lupus nephritis. One of the most studied pathways is the Warburg effect, which is characterized by the preferential use of glycolysis over oxidative phosphorylation in immune cells [56]. The upregulation of glycolysis is thought to be driven by various signaling pathways, including the phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway, the HIF pathway, and the JAK/STAT pathway [57][58]. These pathways have been shown to contribute to the activation of immune cells and the production of autoantibodies in lupus nephritis [58][59][60][61].
The activation of the NLRP3 inflammasome is another key pathway involved in the pathogenesis of lupus nephritis, with the subsequent production of cytokines. The NLRP3 inflammasome contribute to tissue damage in lupus nephritis [62]. Studies have shown that the NLRP3 inflammasome is upregulated in lupus nephritis patients and that its inhibition can ameliorate kidney injury in animal models of lupus nephritis [62][63]. Moreover, dysregulated lipid metabolism has also been implicated in the pathogenesis of lupus nephritis. Studies have shown that increased levels of FFAs can contribute to the activation of immune cells and the production of autoantibodies in lupus nephritis [64][65]. The dysregulation of cholesterol metabolism has also been linked to the development of lupus nephritis. In addition to the above-mentioned pathways, other genes and pathways involved in the dysregulated metabolism in immune cells in lupus nephritis include TLRs, NF-κB, and the IFN pathway. TLRs are involved in the recognition of PAMPs and DAMPs, and their dysregulation has been linked to the activation of immune cells in lupus nephritis [66][67][68]. NF-κB is a transcription factor that regulates the expression of genes involved in inflammation and immune cell activation, and its dysregulation has been shown to contribute to the development of lupus nephritis [69][70]. The type I IFN pathway is another important pathway involved in the activation of immune cells in lupus nephritis, as the overexpression of type I IFN-inducible genes has been observed in lupus nephritis patients [71][72][73].

2.2.2. Diabetic Kidney Disease

Diabetic kidney disease is a common complication of diabetes mellitus and a leading cause of end-stage renal disease [74][75][76]. Dysregulated metabolism and inflammation are key factors in the pathogenesis of diabetic kidney disease. Impaired glucose metabolism leads to the accumulation of AGEs in the kidneys, which contribute to renal dysfunction and fibrosis [77]. GLUT1 and HIF-1α are two genes that have been implicated in the dysregulation of glucose metabolism in immune cells in the context of diabetic kidney disease [15][78].
In addition to dysregulated glucose metabolism, dysregulated lipid metabolism in immune cells has also been implicated in the pathogenesis of diabetic kidney disease. Studies have shown that increased levels of FFAs can contribute to the development of diabetic kidney disease by activating inflammatory pathways and inducing oxidative stress [79][80]. In particular, the peroxisome proliferator-activated receptor (PPAR) family of genes, which regulates lipid metabolism, has been shown to play a role in the pathogenesis of diabetic kidney disease [81][82]. The activation of the NLRP3 inflammasome and subsequent production of pro-inflammatory cytokines have been identified as critical drivers of diabetic kidney disease. The NLRP3 inflammasome is a multiprotein complex involved in the processing and secretion of pro-inflammatory cytokines, and its activation has been implicated in the development of diabetic kidney disease [83]. The inflammasome is activated by a variety of stimuli, including high glucose levels and the accumulation of AGEs [84]. The JAK/STAT signaling pathway is involved in many biological processes, including immune responses and inflammation, and has been implicated in the pathogenesis of diabetic kidney disease [85][86][87][88][89][90][91]. Studies have shown that the JAK/STAT pathway is activated in response to pro-inflammatory cytokines and growth factors, and its dysregulation can contribute to the progression of diabetic kidney disease [92]. The suppressor of cytokine signaling (SOCS) family of genes, which negatively regulates JAK/STAT signaling, has been shown to play a role in the development of diabetic kidney disease [93][94].
In conclusion, dysregulated metabolism and inflammation contribute to the development and progression of diabetic kidney disease through various pathways and genes, including dysregulated glucose and lipid metabolism, activation of the NLRP3 inflammasome, and dysregulated JAK/STAT signaling. Further research in this area may provide novel insights into the mechanisms underlying the development of diabetic kidney disease and help identify new therapeutic targets for the treatment of this condition.

2.2.3. Polycystic Kidney Disease (PKD)

Immunometabolic dysfunction plays a critical role in the pathogenesis of PKD. Dysregulated metabolism in immune cells, such as the activation of the Warburg effect, has been implicated in the development and progression of PKD [95][96]. Additionally, studies have shown that immune cells in PKD exhibit increased mitochondrial stress and metabolic alterations, leading to impaired cellular energetics and increased oxidative stress [97].
One recent study has found that the inflammasome pathway, specifically the NLRP3 inflammasome, is activated in PKD, leading to the production of pro-inflammatory cytokines and subsequent cyst growth [98][99]. The activation of the NLRP3 inflammasome has been linked to the accumulation of damaged mitochondria and the release of mitochondrial DNA, which can trigger an inflammatory response in the kidney [100]. Another study has shown that PKD is associated with altered immune cell metabolism and an increased production of ROS. The authors suggest that this metabolic dysfunction may contribute to the activation of the NLRP3 inflammasome and the subsequent production of pro-inflammatory cytokines in PKD [98][101]. Furthermore, recent research has also linked PKD to dysregulated lipid metabolism in immune cells [36]. One study found that PKD is associated with altered lipid metabolism in T cells, leading to increased T-cell activation and subsequent inflammation in the kidney [102].
In summary, immunometabolic dysfunction, including dysregulated metabolism in immune cells, activation of the inflammasome pathway, altered mitochondrial function, and dysregulated lipid metabolism, contributes to the pathogenesis of PKD. These findings suggest that targeting immunometabolic pathways may provide a potential therapeutic strategy for PKD.

2.2.4. Impact of Immunometabolic Dysregulation on Kidney Transplant Outcomes

Immunometabolic dysregulation has been increasingly recognized as an important contributor to kidney transplant outcomes. The immune response after kidney transplantation involves both the innate and adaptive immune systems, which interact with each other to establish a balance between tolerance and rejection [103][104]. Dysregulated metabolism and inflammation can disrupt this balance, leading to poor transplant outcomes, such as rejection, infection, and chronic allograft dysfunction [105][106].
One key pathway involved in immunometabolic dysregulation after kidney transplantation is the activation of the NLRP3 inflammasome. Studies have shown that activation of the NLRP3 inflammasome in both donor and recipient cells can contribute to the development of acute and chronic rejection [107]. Furthermore, activation of the NLRP3 inflammasome has also been implicated in the development of ischemia–reperfusion injury, a common complication during kidney transplantation [108][109][110]. Dysregulated metabolism in immune cells has also been implicated in poor kidney transplant outcomes. Specifically, the Warburg effect, a phenomenon where immune cells preferentially use glycolysis for energy production instead of oxidative phosphorylation, has been observed in both donor and recipient cells after kidney transplantation [111][112]. This metabolic switch has been associated with increased inflammation and oxidative stress, which can lead to allograft injury and rejection [113][114]. Finally, dysregulation of lipid metabolism in immune cells has also been implicated in poor kidney transplant outcomes [115]. Studies have shown that high levels of triglycerides and low levels of high-density lipoprotein (HDL) cholesterol are associated with an increased risk of acute rejection and chronic allograft dysfunction [116][117]. Dysregulated lipid metabolism in immune cells can also lead to the production of pro-inflammatory cytokines and the activation of the NLRP3 inflammasome [118][119].
In conclusion, immunometabolic dysregulation plays a critical role in kidney transplant outcomes. Dysregulated metabolism and inflammation can disrupt the delicate balance between tolerance and rejection, leading to poor transplant outcomes, such as rejection, infection, and chronic allograft dysfunction. Understanding the mechanisms underlying immunometabolic dysregulation in kidney transplantation may lead to the development of novel therapeutic strategies to improve transplant outcomes.

References

  1. Kovesdy, C.P. Epidemiology of chronic kidney disease: An update 2022. Kidney Int. Suppl. 2022, 12, 7–11.
  2. GBD Chronic Kidney Disease Collaboration. Global, regional, and national burden of chronic kidney disease, 1990–2017: A systematic analysis for the global burden of disease study 2017. Lancet 2020, 395, 709–733.
  3. Chi, H. Immunometabolism at the intersection of metabolic signaling, cell fate, and systems immunology. Cell. Mol. Immunol. 2022, 19, 299–302.
  4. Basso, P.J.; Andrade-Oliveira, V.; Camara, N.O.S. Targeting immune cell metabolism in kidney diseases. Nat. Rev. Nephrol. 2021, 17, 465–480.
  5. Matz, A.J.; Qu, L.; Karlinsey, K.; Vella, A.T.; Zhou, B. Capturing the multifaceted function of adipose tissue macrophages. Front. Immunol. 2023, 14, 1148188.
  6. Qu, L.; Matz, A.J.; Karlinsey, K.; Cao, Z.; Vella, A.T.; Zhou, B. Macrophages at the crossroad of meta-inflammation and inflammaging. Genes 2022, 13, 2074.
  7. Matz, A.J.; Qu, L.; Karlinsey, K.; Zhou, B. Microrna-regulated b cells in obesity. Immunometabolism 2022, 4, e00005.
  8. Makris, K.; Spanou, L. Acute kidney injury: Definition, pathophysiology and clinical phenotypes. Clin. Biochemist. Rev. 2016, 37, 85–98.
  9. Jin, X.; An, C.; Jiao, B.; Safirstein, R.L.; Wang, Y. Amp-activated protein kinase contributes to cisplatin-induced renal epithelial cell apoptosis and acute kidney injury. Am. J. Physiol. Ren. Physiol. 2020, 319, F1073–F1080.
  10. Han, Z.; Ma, K.; Tao, H.; Liu, H.; Zhang, J.; Sai, X.; Li, Y.; Chi, M.; Nian, Q.; Song, L.; et al. A deep insight into regulatory t cell metabolism in renal disease: Facts and perspectives. Front. Immunol. 2022, 13, 826732.
  11. LaFavers, K. Disruption of kidney-immune system crosstalk in sepsis with acute kidney injury: Lessons learned from animal models and their application to human health. Int. J. Mol. Sci. 2022, 23, 1702.
  12. Liu, H.; Li, Y.; Xiong, J. The role of hypoxia-inducible factor-1 alpha in renal disease. Molecules 2022, 27, 7318.
  13. Fu, Z.J.; Wang, Z.Y.; Xu, L.; Chen, X.H.; Li, X.X.; Liao, W.T.; Ma, H.K.; Jiang, M.D.; Xu, T.T.; Xu, J.; et al. Hif-1alpha-bnip3-mediated mitophagy in tubular cells protects against renal ischemia/reperfusion injury. Redox Biol. 2020, 36, 101671.
  14. McGettrick, A.F.; O’Neill, L.A.J. The role of hif in immunity and inflammation. Cell Metab. 2020, 32, 524–536.
  15. Shu, S.; Wang, Y.; Zheng, M.; Liu, Z.; Cai, J.; Tang, C.; Dong, Z. Hypoxia and hypoxia-inducible factors in kidney injury and repair. Cells 2019, 8, 207.
  16. Wang, Z.; Zhang, W. The crosstalk between hypoxia-inducible factor-1alpha and micrornas in acute kidney injury. Exp. Biol. Med. 2020, 245, 427–436.
  17. Taylor, C.T.; Scholz, C.C. The effect of hif on metabolism and immunity. Nat. Rev. Nephrol. 2022, 18, 573–587.
  18. Li, Z.L.; Ji, J.L.; Wen, Y.; Cao, J.Y.; Kharbuja, N.; Ni, W.J.; Yin, D.; Feng, S.T.; Liu, H.; Lv, L.L.; et al. Hif-1alpha is transcriptionally regulated by nf-kappab in acute kidney injury. Am. J. Physiol. Ren. Physiol. 2021, 321, F225–F235.
  19. Li, Z.; Li, N. Epigenetic modification drives acute kidney injury-to-chronic kidney disease progression. Nephron 2021, 145, 737–747.
  20. Tanemoto, F.; Mimura, I. Therapies targeting epigenetic alterations in acute kidney injury-to-chronic kidney disease transition. Pharmaceuticals 2022, 15, 123.
  21. Mimura, I.; Hirakawa, Y.; Kanki, Y.; Kushida, N.; Nakaki, R.; Suzuki, Y.; Tanaka, T.; Aburatani, H.; Nangaku, M. Novel lnc rna regulated by hif-1 inhibits apoptotic cell death in the renal tubular epithelial cells under hypoxia. Physiol. Rep. 2017, 5, e13203.
  22. Forstermann, U.; Sessa, W.C. Nitric oxide synthases: Regulation and function. Eur. Heart J. 2012, 33, 829–837.
  23. Oliveira, F.; Assreuy, J.; Sordi, R. The role of nitric oxide in sepsis-associated kidney injury. Biosci. Rep. 2022, 42, BSR20220093.
  24. Wang, J.; Cong, X.; Miao, M.; Yang, Y.; Zhang, J. Inhaled nitric oxide and acute kidney injury risk: A meta-analysis of randomized controlled trials. Ren. Fail. 2021, 43, 281–290.
  25. Carlstrom, M. Nitric oxide signalling in kidney regulation and cardiometabolic health. Nat. Rev. Nephrol. 2021, 17, 575–590.
  26. Ludes, P.O.; de Roquetaillade, C.; Chousterman, B.G.; Pottecher, J.; Mebazaa, A. Role of damage-associated molecular patterns in septic acute kidney injury, from injury to recovery. Front. Immunol. 2021, 12, 606622.
  27. Meissner, M.; Viehmann, S.F.; Kurts, C. Dampening sterile inflammation of the kidney. Kidney Int. 2019, 95, 489–491.
  28. Vazquez-Carballo, C.; Guerrero-Hue, M.; Garcia-Caballero, C.; Rayego-Mateos, S.; Opazo-Rios, L.; Morgado-Pascual, J.L.; Herencia-Bellido, C.; Vallejo-Mudarra, M.; Cortegano, I.; Gaspar, M.L.; et al. Toll-like receptors in acute kidney injury. Int. J. Mol. Sci. 2021, 22, 816.
  29. Liu, C.; Shen, Y.; Huang, L.; Wang, J. Tlr2/caspase-5/panx1 pathway mediates necrosis-induced nlrp3 inflammasome activation in macrophages during acute kidney injury. Cell Death Discov. 2022, 8, 232.
  30. Lin, Q.; Li, S.; Jiang, N.; Jin, H.; Shao, X.; Zhu, X.; Wu, J.; Zhang, M.; Zhang, Z.; Shen, J.; et al. Inhibiting nlrp3 inflammasome attenuates apoptosis in contrast-induced acute kidney injury through the upregulation of hif1a and bnip3-mediated mitophagy. Autophagy 2021, 17, 2975–2990.
  31. Akhter, J.; Khan, J.; Baghel, M.; Beg, M.M.A.; Goswami, P.; Afjal, M.A.; Ahmad, S.; Habib, H.; Najmi, A.K.; Raisuddin, S. Nlrp3 inflammasome in rosmarinic acid-afforded attenuation of acute kidney injury in mice. Sci. Rep. 2022, 12, 1313.
  32. Kim, H.J.; Lee, D.W.; Ravichandran, K.; Keys, D.O.; Akcay, A.; Nguyen, Q.; He, Z.; Jani, A.; Ljubanovic, D.; Edelstein, C.L. Nlrp3 inflammasome knockout mice are protected against ischemic but not cisplatin-induced acute kidney injury. J. Pharmacol. Exp. Ther. 2013, 346, 465–472.
  33. Li, J.; Yang, Y.; Wang, Y.; Li, Q.; He, F. Metabolic signatures of immune cells in chronic kidney disease. Expert Rev. Mol. Med. 2022, 24, e40.
  34. de Boer, I.H. Vitamin d and glucose metabolism in chronic kidney disease. Curr. Opin. Nephrol. Hypertens. 2008, 17, 566–572.
  35. Gupta, N.; Wish, J.B. Hypoxia-inducible factor prolyl hydroxylase inhibitors: A potential new treatment for anemia in patients with ckd. Am. J. Kidney Dis. Off. J. Natl. Kidney Found. 2017, 69, 815–826.
  36. Irazabal, M.V.; Torres, V.E. Reactive oxygen species and redox signaling in chronic kidney disease. Cells 2020, 9, 1342.
  37. Wei, X.; Hou, Y.; Long, M.; Jiang, L.; Du, Y. Molecular mechanisms underlying the role of hypoxia-inducible factor-1 alpha in metabolic reprogramming in renal fibrosis. Front. Endocrinol. 2022, 13, 927329.
  38. Mokas, S.; Lariviere, R.; Lamalice, L.; Gobeil, S.; Cornfield, D.N.; Agharazii, M.; Richard, D.E. Hypoxia-inducible factor-1 plays a role in phosphate-induced vascular smooth muscle cell calcification. Kidney Int. 2016, 90, 598–609.
  39. Vilaysane, A.; Chun, J.; Seamone, M.E.; Wang, W.; Chin, R.; Hirota, S.; Li, Y.; Clark, S.A.; Tschopp, J.; Trpkov, K.; et al. The nlrp3 inflammasome promotes renal inflammation and contributes to ckd. J. Am. Soc. Nephrol. 2010, 21, 1732–1744.
  40. Gai, Z.; Wang, T.; Visentin, M.; Kullak-Ublick, G.A.; Fu, X.; Wang, Z. Lipid accumulation and chronic kidney disease. Nutrients 2019, 11, 722.
  41. Pei, K.; Gui, T.; Li, C.; Zhang, Q.; Feng, H.; Li, Y.; Wu, J.; Gai, Z. Recent progress on lipid intake and chronic kidney disease. BioMed Res. Int. 2020, 2020, 3680397.
  42. Sepe, V.; Libetta, C.; Gregorini, M.; Rampino, T. The innate immune system in human kidney inflammaging. J. Nephrol. 2022, 35, 381–395.
  43. Zewinger, S.; Schumann, T.; Fliser, D.; Speer, T. Innate immunity in ckd-associated vascular diseases. Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc.-Eur. Ren. Assoc. 2016, 31, 1813–1821.
  44. Lee, H.; Fessler, M.B.; Qu, P.; Heymann, J.; Kopp, J.B. Macrophage polarization in innate immune responses contributing to pathogenesis of chronic kidney disease. BMC Nephrol. 2020, 21, 270.
  45. Zhang, H.; Sun, S.C. Nf-kappab in inflammation and renal diseases. Cell Biosci. 2015, 5, 63.
  46. Rangan, G.; Wang, Y.; Harris, D. Nf-kappab signalling in chronic kidney disease. Front. Biosci. 2009, 14, 3496–3522.
  47. Huang, G.; Zhang, Y.; Zhang, Y.; Ma, Y. Chronic kidney disease and nlrp3 inflammasome: Pathogenesis, development and targeted therapeutic strategies. Biochem. Biophys. Rep. 2023, 33, 101417.
  48. Zhang, H.; Wang, Z. Effect and regulation of the nlrp3 inflammasome during renal fibrosis. Front. Cell Dev. Biol. 2019, 7, 379.
  49. Siragy, H.M.; Carey, R.M. Role of the intrarenal renin-angiotensin-aldosterone system in chronic kidney disease. Am. J. Nephrol. 2010, 31, 541–550.
  50. Gaudreault-Tremblay, M.M.; Foster, B.J. Benefits of continuing raas inhibitors in advanced ckd. Clin. J. Am. Soc. Nephrol. 2020, 15, 592–593.
  51. Remuzzi, G.; Perico, N.; Macia, M.; Ruggenenti, P. The role of renin-angiotensin-aldosterone system in the progression of chronic kidney disease. Kidney Int. Suppl. 2005, 68, S57–S65.
  52. Lazar, S.; Kahlenberg, J.M. Systemic lupus erythematosus: New diagnostic and therapeutic approaches. Annu. Rev. Med. 2023, 74, 339–352.
  53. Ma, L.; Roach, T.; Morel, L. Immunometabolic alterations in lupus: Where do they come from and where do we go from there? Curr. Opin. Immunol. 2022, 78, 102245.
  54. Liu, X.; Du, H.; Sun, Y.; Shao, L. Role of abnormal energy metabolism in the progression of chronic kidney disease and drug intervention. Ren. Fail. 2022, 44, 790–805.
  55. Fornoni, A.; Merscher, S. Lipid metabolism gets in a jaml during kidney disease. Cell Metab. 2020, 32, 903–905.
  56. Liberti, M.V.; Locasale, J.W. The warburg effect: How does it benefit cancer cells? Trends Biochem. Sci. 2016, 41, 211–218.
  57. Sun, Q.; Chen, X.; Ma, J.; Peng, H.; Wang, F.; Zha, X.; Wang, Y.; Jing, Y.; Yang, H.; Chen, R.; et al. Mammalian target of rapamycin up-regulation of pyruvate kinase isoenzyme type m2 is critical for aerobic glycolysis and tumor growth. Proc. Natl. Acad. Sci. USA 2011, 108, 4129–4134.
  58. Chen, H.; Liu, N.; Zhuang, S. Macrophages in renal injury, repair, fibrosis following acute kidney injury and targeted therapy. Front. Immunol. 2022, 13, 934299.
  59. Scuron, M.D.; Fay, B.L.; Connell, A.J.; Oliver, J.; Smith, P.A. The pi3kdelta inhibitor parsaclisib ameliorates pathology and reduces autoantibody formation in preclinical models of systemic lupus erythematosus and sjögren’s syndrome. Int. Immunopharmacol. 2021, 98, 107904.
  60. Ripoll, E.; de Ramon, L.; Draibe Bordignon, J.; Merino, A.; Bolanos, N.; Goma, M.; Cruzado, J.M.; Grinyo, J.M.; Torras, J. Jak3-stat pathway blocking benefits in experimental lupus nephritis. Arthritis Res. Ther. 2016, 18, 134.
  61. Zhao, W.; Wu, C.; Li, L.J.; Fan, Y.G.; Pan, H.F.; Tao, J.H.; Leng, R.X.; Ye, D.Q. Rnai silencing of hif-1alpha ameliorates lupus development in mrl/lpr mice. Inflammation 2018, 41, 1717–1730.
  62. Oliveira, C.B.; Lima, C.A.D.; Vajgel, G.; Sandrin-Garcia, P. The role of nlrp3 inflammasome in lupus nephritis. Int. J. Mol. Sci. 2021, 22, 12476.
  63. Wu, D.; Ai, L.; Sun, Y.; Yang, B.; Chen, S.; Wang, Q.; Kuang, H. Role of nlrp3 inflammasome in lupus nephritis and therapeutic targeting by phytochemicals. Front. Pharmacol. 2021, 12, 621300.
  64. Pestka, J.J.; Akbari, P.; Wierenga, K.A.; Bates, M.A.; Gilley, K.N.; Wagner, J.G.; Lewandowski, R.P.; Rajasinghe, L.D.; Chauhan, P.S.; Lock, A.L.; et al. Omega-3 polyunsaturated fatty acid intervention against established autoimmunity in a murine model of toxicant-triggered lupus. Front. Immunol. 2021, 12, 653464.
  65. Pestka, J.J.; Vines, L.L.; Bates, M.A.; He, K.; Langohr, I. Comparative effects of n-3, n-6 and n-9 unsaturated fatty acid-rich diet consumption on lupus nephritis, autoantibody production and cd4+ t cell-related gene responses in the autoimmune nzbwf1 mouse. PLoS ONE 2014, 9, e100255.
  66. Wolf, S.J.; Theros, J.; Reed, T.J.; Liu, J.; Grigorova, I.L.; Martinez-Colon, G.; Jacob, C.O.; Hodgin, J.B.; Kahlenberg, J.M. Tlr7-mediated lupus nephritis is independent of type i ifn signaling. J. Immunol. 2018, 201, 393–405.
  67. Ding, X.; Ren, Y.; He, X. Ifn-i mediates lupus nephritis from the beginning to renal fibrosis. Front. Immunol. 2021, 12, 676082.
  68. Devarapu, S.K.; Anders, H.J. Toll-like receptors in lupus nephritis. J. Biomed. Sci. 2018, 25, 35.
  69. He, L.Y.; Niu, S.Q.; Yang, C.X.; Tang, P.; Fu, J.J.; Tan, L.; Li, Y.; Hua, Y.N.; Liu, S.J.; Guo, J.L. Cordyceps proteins alleviate lupus nephritis through modulation of the stat3/mtor/nf-small ka, cyrillicb signaling pathway. J. Ethnopharmacol. 2023, 309, 116284.
  70. Zou, L.; Sun, L.; Hua, R.; Wu, Y.; Sun, L.; Chen, T. Degradation of ubiquitin-editing enzyme a20 following autophagy activation promotes rnf168 nuclear translocation and nf-kappab activation in lupus nephritis. J. Innate Immun. 2023, 15, 428–441.
  71. Karasawa, T.; Sato, R.; Imaizumi, T.; Hashimoto, S.; Fujita, M.; Aizawa, T.; Tsugawa, K.; Kawaguchi, S.; Seya, K.; Terui, K.; et al. Glomerular endothelial expression of type i ifn-stimulated gene, dexd/h-box helicase 60 via toll-like receptor 3 signaling: Possible involvement in the pathogenesis of lupus nephritis. Ren. Fail. 2022, 44, 137–145.
  72. Dunlap, G.S.; Billi, A.C.; Xing, X.; Ma, F.; Maz, M.P.; Tsoi, L.C.; Wasikowski, R.; Hodgin, J.B.; Gudjonsson, J.E.; Kahlenberg, J.M.; et al. Single-cell transcriptomics reveals distinct effector profiles of infiltrating t cells in lupus skin and kidney. JCI Insight 2022, 7, e156341.
  73. Tsao, Y.P.; Tseng, F.Y.; Chao, C.W.; Chen, M.H.; Yeh, Y.C.; Abdulkareem, B.O.; Chen, S.Y.; Chuang, W.T.; Chang, P.C.; Chen, I.C.; et al. Nlrp12 is an innate immune checkpoint for repressing ifn signatures and attenuating lupus nephritis progression. J. Clin. Investig. 2023, 133, e157272.
  74. Sagoo, M.K.; Gnudi, L. Diabetic nephropathy: An overview. Methods Mol. Biol. 2020, 2067, 3–7.
  75. Wang, X.; Zhao, L.; Ajay, A.K.; Jiao, B.; Zhang, X.; Wang, C.; Gao, X.; Yuan, Z.; Liu, H.; Liu, W.J. Qiditangshen granules activate renal nutrient-sensing associated autophagy in db/db mice. Front. Physiol. 2019, 10, 1224.
  76. Han, H.; Wang, L.; Du, H.; Jiang, J.; Hu, C.; Zhang, G.; Liu, S.; Zhang, X.; Liu, T.; Hu, S. Expedited biliopancreatic juice flow to the distal gut benefits the diabetes control after duodenal-jejunal bypass. Obes. Surg. 2015, 25, 1802–1809.
  77. Sanajou, D.; Ghorbani Haghjo, A.; Argani, H.; Aslani, S. Age-rage axis blockade in diabetic nephropathy: Current status and future directions. Eur. J. Pharmacol. 2018, 833, 158–164.
  78. Kobayashi, H.; Gilbert, V.; Liu, Q.; Kapitsinou, P.P.; Unger, T.L.; Rha, J.; Rivella, S.; Schlondorff, D.; Haase, V.H. Myeloid cell-derived hypoxia-inducible factor attenuates inflammation in unilateral ureteral obstruction-induced kidney injury. J. Immunol. 2012, 188, 5106–5115.
  79. Tsai, I.T.; Wu, C.C.; Hung, W.C.; Lee, T.L.; Hsuan, C.F.; Wei, C.T.; Lu, Y.C.; Yu, T.H.; Chung, F.M.; Lee, Y.J.; et al. Fabp1 and fabp2 as markers of diabetic nephropathy. Int. J. Med. Sci. 2020, 17, 2338–2345.
  80. Sieber, J.; Jehle, A.W. Free fatty acids and their metabolism affect function and survival of podocytes. Front. Endocrinol. 2014, 5, 186.
  81. Tomita, Y.; Lee, D.; Tsubota, K.; Kurihara, T. Pparalpha agonist oral therapy in diabetic retinopathy. Biomedicines 2020, 8, 433.
  82. Hu, Y.; Chen, Y.; Ding, L.; He, X.; Takahashi, Y.; Gao, Y.; Shen, W.; Cheng, R.; Chen, Q.; Qi, X.; et al. Pathogenic role of diabetes-induced ppar-alpha down-regulation in microvascular dysfunction. Proc. Natl. Acad. Sci. USA 2013, 110, 15401–15406.
  83. Ding, S.; Xu, S.; Ma, Y.; Liu, G.; Jang, H.; Fang, J. Modulatory mechanisms of the nlrp3 inflammasomes in diabetes. Biomolecules 2019, 9, 850.
  84. Wan, L.; Bai, X.; Zhou, Q.; Chen, C.; Wang, H.; Liu, T.; Xue, J.; Wei, C.; Xie, L. The advanced glycation end-products (ages)/ros/nlrp3 inflammasome axis contributes to delayed diabetic corneal wound healing and nerve regeneration. Int. J. Biol. Sci. 2022, 18, 809–825.
  85. Shi, X.; Jiao, B.; Chen, Y.; Li, S.; Chen, L. Mxa is a positive regulator of type i ifn signaling in hcv infection. J. Med. Virol. 2017, 89, 2173–2180.
  86. Chen, Y.; Jiao, B.; Yao, M.; Shi, X.; Zheng, Z.; Li, S.; Chen, L. Isg12a inhibits hcv replication and potentiates the anti-hcv activity of ifn-alpha through activation of the jak/stat signaling pathway independent of autophagy and apoptosis. Virus Res. 2017, 227, 231–239.
  87. Duan, X.; Li, S.; Holmes, J.A.; Tu, Z.; Li, Y.; Cai, D.; Liu, X.; Li, W.; Yang, C.; Jiao, B.; et al. Microrna 130a regulates both hepatitis c virus and hepatitis b virus replication through a central metabolic pathway. J. Virol. 2018, 92, e02009-17.
  88. Chen, X.; Ye, H.; Li, S.; Jiao, B.; Wu, J.; Zeng, P.; Chen, L. Severe fever with thrombocytopenia syndrome virus inhibits exogenous type i ifn signaling pathway through its nss invitro. PLoS ONE 2017, 12, e0172744.
  89. Li, Y.; Li, S.; Duan, X.; Chen, Y.; Jiao, B.; Ye, H.; Yao, M.; Chen, L. Interferon-stimulated gene 15 conjugation stimulates hepatitis b virus production independent of type i interferon signaling pathway in vitro. Mediat. Inflamm. 2016, 2016, 7417648.
  90. Jiao, B.; Shi, X.; Chen, Y.; Ye, H.; Yao, M.; Hong, W.; Li, S.; Duan, X.; Li, Y.; Wang, Y.; et al. Insulin receptor substrate-4 interacts with ubiquitin-specific protease 18 to activate the jak/stat signaling pathway. Oncotarget 2017, 8, 105923–105935.
  91. Yuan, Y.; Jiao, B.; Qu, L.; Yang, D.; Liu, R. The development of covid-19 treatment. Front. Immunol. 2023, 14, 1125246.
  92. Chen, D.; Liu, Y.; Chen, J.; Lin, H.; Guo, H.; Wu, Y.; Xu, Y.; Zhou, Y.; Zhou, W.; Lu, R.; et al. Jak/stat pathway promotes the progression of diabetic kidney disease via autophagy in podocytes. Eur. J. Pharmacol. 2021, 902, 174121.
  93. Lv, L.L.; Feng, Y.; Wu, M.; Wang, B.; Li, Z.L.; Zhong, X.; Wu, W.J.; Chen, J.; Ni, H.F.; Tang, T.T.; et al. Exosomal mirna-19b-3p of tubular epithelial cells promotes m1 macrophage activation in kidney injury. Cell Death Differ. 2020, 27, 210–226.
  94. Zhu, M.; Wang, H.; Chen, J.; Zhu, H. Sinomenine improve diabetic nephropathy by inhibiting fibrosis and regulating the jak2/stat3/socs1 pathway in streptozotocin-induced diabetic rats. Life Sci. 2021, 265, 118855.
  95. Riwanto, M.; Kapoor, S.; Rodriguez, D.; Edenhofer, I.; Segerer, S.; Wuthrich, R.P. Inhibition of aerobic glycolysis attenuates disease progression in polycystic kidney disease. PLoS ONE 2016, 11, e0146654.
  96. Podrini, C.; Cassina, L.; Boletta, A. Metabolic reprogramming and the role of mitochondria in polycystic kidney disease. Cell. Signal. 2020, 67, 109495.
  97. Nguyen, D.T.; Kleczko, E.K.; Dwivedi, N.; Monaghan, M.T.; Gitomer, B.Y.; Chonchol, M.B.; Clambey, E.T.; Nemenoff, R.A.; Klawitter, J.; Hopp, K. The tryptophan-metabolizing enzyme indoleamine 2,3-dioxygenase 1 regulates polycystic kidney disease progression. JCI Insight 2023, 8.
  98. Swenson-Fields, K.I.; Ward, C.J.; Lopez, M.E.; Fross, S.; Heimes Dillon, A.L.; Meisenheimer, J.D.; Rabbani, A.J.; Wedlock, E.; Basu, M.K.; Jansson, K.P.; et al. Caspase-1 and the inflammasome promote polycystic kidney disease progression. Front. Mol. Biosci. 2022, 9, 971219.
  99. Raptis, V.; Loutradis, C.; Boutou, A.K.; Faitatzidou, D.; Sioulis, A.; Ferro, C.J.; Papagianni, A.; Sarafidis, P.A. Serum copeptin, nlpr3, and supar levels among patients with autosomal-dominant polycystic kidney disease with and without impaired renal function. Cardiorenal Med. 2020, 10, 440–451.
  100. Xiong, W.; Meng, X.F.; Zhang, C. Nlrp3 inflammasome in metabolic-associated kidney diseases: An update. Front. Immunol. 2021, 12, 714340.
  101. Granata, S.; Masola, V.; Zoratti, E.; Scupoli, M.T.; Baruzzi, A.; Messa, M.; Sallustio, F.; Gesualdo, L.; Lupo, A.; Zaza, G. Nlrp3 inflammasome activation in dialyzed chronic kidney disease patients. PLoS ONE 2015, 10, e0122272.
  102. Kleczko, E.K.; Marsh, K.H.; Tyler, L.C.; Furgeson, S.B.; Bullock, B.L.; Altmann, C.J.; Miyazaki, M.; Gitomer, B.Y.; Harris, P.C.; Weiser-Evans, M.C.M.; et al. Cd8(+) t cells modulate autosomal dominant polycystic kidney disease progression. Kidney Int. 2018, 94, 1127–1140.
  103. Huang, D.L.; He, Y.R.; Liu, Y.J.; He, H.Y.; Gu, Z.Y.; Liu, Y.M.; Liu, W.J.; Luo, Z.; Ju, M.J. The immunomodulation role of th17 and treg in renal transplantation. Front. Immunol. 2023, 14, 1113560.
  104. Tanimine, N.; Turka, L.A.; Priyadharshini, B. Navigating t-cell immunometabolism in transplantation. Transplantation 2018, 102, 230–239.
  105. Tran, D.T.; Sundararaj, K.; Atkinson, C.; Nadig, S.N. T-cell immunometabolism: Therapeutic implications in organ transplantation. Transplantation 2021, 105, e191–e201.
  106. Kazmi, S.; Khan, M.A.; Shamma, T.; Altuhami, A.; Assiri, A.M.; Broering, D.C. Therapeutic nexus of t cell immunometabolism in improving transplantation immunotherapy. Int. Immunopharmacol. 2022, 106, 108621.
  107. Lucas-Ruiz, F.; Penin-Franch, A.; Pons, J.A.; Ramirez, P.; Pelegrin, P.; Cuevas, S.; Baroja-Mazo, A. Emerging role of nlrp3 inflammasome and pyroptosis in liver transplantation. Int. J. Mol. Sci. 2022, 23, 14396.
  108. Wang, M.; Pan, W.; Xu, Y.; Zhang, J.; Wan, J.; Jiang, H. Microglia-mediated neuroinflammation: A potential target for the treatment of cardiovascular diseases. J. Inflamm. Res. 2022, 15, 3083–3094.
  109. Su, X.; Liu, B.; Wang, S.; Wang, Y.; Zhang, Z.; Zhou, H.; Li, F. Nlrp3 inflammasome: A potential therapeutic target to minimize renal ischemia/reperfusion injury during transplantation. Transpl. Immunol. 2022, 75, 101718.
  110. Liu, Y.; Lei, Z.; Chai, H.; Kang, Q.; Qin, X. Salidroside alleviates hepatic ischemia-reperfusion injury during liver transplant in rat through regulating tlr-4/nf-kappab/nlrp3 inflammatory pathway. Sci. Rep. 2022, 12, 13973.
  111. Hecking, M.; Kainz, A.; Werzowa, J.; Haidinger, M.; Doller, D.; Tura, A.; Karaboyas, A.; Horl, W.H.; Wolzt, M.; Sharif, A.; et al. Glucose metabolism after renal transplantation. Diabetes Care 2013, 36, 2763–2771.
  112. Baker, R.J.; Marks, S.D. Management of chronic renal allograft dysfunction and when to re-transplant. Pediatr. Nephrol. 2019, 34, 599–603.
  113. Nafar, M.; Sahraei, Z.; Salamzadeh, J.; Samavat, S.; Vaziri, N.D. Oxidative stress in kidney transplantation: Causes, consequences, and potential treatment. Iran. J. Kidney Dis. 2011, 5, 357–372.
  114. Diaz-De la Cruz, E.N.; Cerrillos-Gutierrez, J.I.; Garcia-Sanchez, A.; Andrade-Sierra, J.; Cardona-Munoz, E.G.; Rojas-Campos, E.; Gonzalez-Espinoza, E.; Miranda-Diaz, A.G. The alteration of pro-inflammatory cytokines and oxidative stress markers at six-month post-living kidney donation. Front. Med. 2020, 7, 382.
  115. Pandya, V.; Rao, A.; Chaudhary, K. Lipid abnormalities in kidney disease and management strategies. World J. Nephrol. 2015, 4, 83–91.
  116. Barn, K.; Laftavi, M.; Pierce, D.; Ying, C.; Boden, W.E.; Pankewycz, O. Low levels of high-density lipoprotein cholesterol: An independent risk factor for late adverse cardiovascular events in renal transplant recipients. Transpl. Int. Off. J. Eur. Soc. Organ Transplant. 2010, 23, 574–579.
  117. Bowe, B.; Xie, Y.; Xian, H.; Balasubramanian, S.; Al-Aly, Z. Low levels of high-density lipoprotein cholesterol increase the risk of incident kidney disease and its progression. Kidney Int. 2016, 89, 886–896.
  118. Meyers, A.K.; Zhu, X. The nlrp3 inflammasome: Metabolic regulation and contribution to inflammaging. Cells 2020, 9, 1808.
  119. Liang, J.J.; Fraser, I.D.C.; Bryant, C.E. Lipid regulation of nlrp3 inflammasome activity through organelle stress. Trends Immunol. 2021, 42, 807–823.
More
Information
Subjects: Others
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 317
Revisions: 2 times (View History)
Update Date: 06 Jul 2023
1000/1000
Video Production Service