Review on the neuron damage parameters in patients with end-stage renal disease

Main Article Content

Basim Abd Al-Raheem Twaij
Hussein Kadhem Al-Hakeim
Mazin Fadhil Altufaili

Keywords

Damage, Parameters, Patients, Protien

Abstract

Many studies have found toxic effects on the body and mental state at near and long-term parameters in end-stage renal disease (ESRD) patients of neuronal integrity. Consequently, studying chemical molecules that cause tissue damage to neurons is essential for understanding the mechanism of toxicity and available treatment prospects. Which includes some parameters of myelin basic protein (MBP), ionized calcium-binding adaptor molecule 1 IBA1, calcium-binding protein B (S100B), Glial fibrillary acidic protein (GFAP), neuroepithelial stem cell protein (Nestin), neurofilament light polypeptide (NFL), neuron-specific enolase, T-tau and claudin proteins.
The published papers on changes in neuronal damage final products in ESRD patients were reviewed, and the explanations obtained from previous research were collected. It is concluded from this review that causes an increase in parameters in patients with ESRD lead to neurodegeneration, which leads to severe damage to patients health that requires therapeutic intervention to reduce the harmful effects of parameters on the health of patients.

Abstract 150 | PDF Downloads 134

References

1. Guo, L., et al., Diuretic resistance in patients with kidney disease: Challenges and opportunities. 2023. 157: p. 114058.
2. Banerjee, D., et al., Management of hypertension and renin-angiotensin-aldosterone system blockade in adults with diabetic kidney disease: Association of British Clinical Diabetologists and the Renal Association UK guideline update 2021. 2022. 23(1): p. 1-31.
3. Aier, A., et al., Psychological aspects in children and parents of children with chronic kidney disease and their families. 2022. 65(5): p. 222.
4. Akhtar, M., et al., Diabetic kidney disease: past and present. 2020. 27(2): p. 87-97.
5. Luciano, R.L. and G.W. Moeckel, Update on the native kidney biopsy: core curriculum 2019. American Journal of Kidney Diseases, 2019. 73(3): p. 404-415.
6. Nesrallah, G.E., et al., Canadian Society of Nephrology 2014 clinical practice guideline for timing the initiation of chronic dialysis. Cmaj, 2014. 186(2): p. 112-117.
7. Barsoum, R.S., Chronic kidney disease in the developing world. New England Journal of Medicine, 2006. 354(10): p. 997-999.
8. Najafi, I., Peritoneal dialysis in Iran and the Middle East. Peritoneal dialysis international, 2009. 29(2_suppl): p. 217-221.
9. Izeidi, P.P., et al., Cost estimate of chronic hemodialysis in Kinshasa, the Democratic Republic of the Congo: A prospective study in two centers. Hemodialysis International, 2020. 24(1): p. 121-128.
10. Asad, H.N., et al., A causal-pathway phenotype of chronic fatigue syndrome due to hemodialysis in patients with end-stage renal disease. 2023. 22(2): p. 191-206.
11. Jwad, S.M. and H.Y.J.R.F. AL-Fatlawi, Diabetic Nephropathy and Its Effect on the Physiological Factors of The body. 2022. 8: p. 10.
12. Mikkelsen, H., et al., Glomerular proteomic profiling of kidney biopsies with hypertensive nephropathy reveals a signature of disease progression. 2023. 46(1): p. 144-156.
13. Lees, J.S., et al., Author Correction: Glomerular filtration rate by differing measures, albuminuria and prediction of cardiovascular disease, mortality and end-stage kidney disease. 2020. 26(8): p. 1308-1308.
14. Brash, J.L., Spinal cord pathology of ovine CLN5 and CLN6 neuronal ceroid lipofuscinoses (Batten disease): A thesis submitted in partial fulfilment of the requirements for the Degree of Master at Lincoln University. 2023, Lincoln University.
15. Pickett, L.A., et al., Microglia phagocytosis mediates the volume and function of the rat sexually dimorphic nucleus of the preoptic area. 2023. 120(10): p. e2212646120.
16. Decourt, B., et al., The cause of Alzheimer’s disease: the theory of multipathology convergence to chronic neuronal stress. 2022. 13(1): p. 37.
17. Lei, F., et al., GSK-3 inhibitor promotes neuronal cell regeneration and functional recovery in a rat model of spinal cord injury. 2019. 2019.
18. Grader, E. and A.J.T.b.i.r.w. Bateman, Introduction to brain anatomy and mechanisms of injury. 2017: p. 15-35.
19. Rahman, M., et al., Emerging Role of Neuron-Glia in Neurological Disorders: At a Glance. 2022. 2022.
20. Vassall, K.A., V.V. Bamm, and G.J.B.J. Harauz, MyelStones: the executive roles of myelin basic protein in myelin assembly and destabilization in multiple sclerosis. 2015. 472(1): p. 17-32.
21. Bigbee, J.W.J.G.o.t.N.S., Cells of the Central Nervous System: An Overview of Their Structure and Function. 2023: p. 41-64.
22. Vassall, K.A., et al., Substitutions mimicking deimination and phosphorylation of 18.5-kDa myelin basic protein exert local structural effects that subtly influence its global folding. 2016. 1858(6): p. 1262-1277.
23. Bando, Y.J.C. and E. Neuroimmunology, Mechanism of demyelination and remyelination in multiple sclerosis. 2020. 11: p. 14-21.
24. Wimmer, I., et al., PECAM-1 stabilizes blood-brain barrier integrity and favors paracellular T-cell diapedesis across the blood-brain barrier during neuroinflammation. 2019. 10: p. 711.
25. Cui, L.-Y., S.-F. Chu, and N.-H.J.I.i. Chen, The role of chemokines and chemokine receptors in multiple sclerosis. 2020. 83: p. 106314.
26. Bromberg, M.B., Peripheral Neuropathies: A Practical Approach. 2018: Cambridge University Press.
27. Stadelmann, C., et al., Myelin in the central nervous system: structure, function, and pathology. 2019. 99(3): p. 1381-1431.
28. Eggermann, K., et al., Hereditary neuropathies: clinical presentation and genetic panel diagnosis. 2018. 115(6): p. 91.
29. Łowiec, P., P. Trzonkowski, and K. Chwojnicki, Multiple sclerosis–new therapeutic directions. 2019.
30. Raasakka, A. and P.J.C. Kursula, How does protein zero assemble compact myelin? 2020. 9(8): p. 1832.
31. Ziegler, V., et al., Mesangial cells regulate the single nephron GFR and preserve the integrity of the glomerular filtration barrier: An intravital multiphoton microscopy study. 2021. 231(4): p. e13592.
32. McDermott, M., et al., Mammalian phospholipase D: Function, and therapeutics. 2020. 78: p. 101018.
33. Ho, Y.-S., et al., Impact of unilateral ureteral obstruction on cognition and neurodegeneration. 2021. 169: p. 112-127.
34. Kambhampati, S.P., et al., Systemic dendrimer nanotherapies for targeted suppression of choroidal inflammation and neovascularization in age-related macular degeneration. 2021. 335: p. 527-540.
35. Dello Russo, C., et al., Exploiting microglial functions for the treatment of glioblastoma. 2017. 17(3): p. 267-281.
36. Shi, F.-J., et al., Is Iba-1 protein expression a sensitive marker for microglia activation in experimental diabetic retinopathy? 2021. 14(2): p. 200.
37. Chib, S., S.J.N. Singh, and Teratology, Manganese and related neurotoxic pathways: A potential therapeutic target in neurodegenerative diseases. 2022: p. 107124.
38. Kadian, M., G. Sharma, and A. Kumar, Iron-Calcium Crosstalk in Neurodegenerative Diseases, in Brain-Iron Cross Talk. 2023, Springer. p. 109-137.
39. Williams, T., D.R. Borchelt, and P.J.M.n. Chakrabarty, Therapeutic approaches targeting Apolipoprotein E function in Alzheimer’s disease. 2020. 15(1): p. 1-19.
40. Wolters, E.C., et al., Naïve BM-derived stem cells (Neuro-Cells) may modify acute and chronic neurodegenerative disorders by modulating macrophage behaviors. 2021. 1(1): p. 3.
41. Tamaru, T., et al., Glial scar survives until the chronic phase by recruiting scar-forming astrocytes after spinal cord injury. 2023. 359: p. 114264.
42. das Neves, S.P., et al., Altered astrocytic function in experimental neuroinflammation and multiple sclerosis. 2021. 69(6): p. 1341-1368.
43. Bonsack, B., et al., Brief overview: Protective roles of astrocyte-derived pentraxin-3 in blood-brain barrier integrity. 2019. 5(3): p. 145.
44. Zhang, W., et al., The blood brain barrier in cerebral ischemic injury–Disruption and repair. 2020. 1(1): p. 34-53.
45. Braun, M., et al., White matter damage after traumatic brain injury: a role for damage associated molecular patterns. 2017. 1863(10): p. 2614-2626.
46. Alpdemir, M., et al., Serum neuron specific enolase and S-100B levels in hemodialysis and peritoneal dialysis patients. 2019. 35(2): p. 83.
47. Liao, G., et al., Antidyslipidemia Pharmacotherapy in Chronic Kidney Disease: A Systematic Review and Bayesian Network Meta-Analysis. 2023. 15(1): p. 6.
48. Hernandez, L., et al., Blood–brain barrier and gut barrier dysfunction in chronic kidney disease with a focus on circulating biomarkers and tight junction proteins. 2022. 12(1): p. 1-14.
49. Kawata, K., R. Tierney, and D.J.H.o.c.n. Langford, Blood and cerebrospinal fluid biomarkers. 2018. 158: p. 217-233.
50. Maciak, K., A. Dziedzic, and J.J.T.F.J. Saluk, Possible role of the NLRP3 inflammasome and the gut–brain axis in multiple sclerosis‐related depression. 2023. 37(1): p. e22687.
51. Zhao, Z., et al., Applying membrane technology in microalgae industry: A comprehensive review. 2023. 172: p. 113041.
52. Lu, T. and J.C.J.B.o.s.D. Mar, Investigating transcriptome-wide sex dimorphism by multi-level analysis of single-cell RNA sequencing data in ten mouse cell types. 2020. 11(1): p. 1-20.
53. Cattane, N., et al., Preclinical animal models of mental illnesses to translate findings from the bench to the bedside: Molecular brain mechanisms and peripheral biomarkers associated to early life stress or immune challenges. 2022. 58: p. 55-79.
54. MacDonald, A.J., et al., Astrocytes in neuroendocrine systems: An overview. 2019. 31(5): p. e12726.
55. Cansell, C., et al., Dietary fat exacerbates postprandial hypothalamic inflammation involving glial fibrillary acidic protein‐positive cells and microglia in male mice. 2021. 69(1): p. 42-60.
56. Hol, E.M. and Y.J.C.S.H.p.i.b. Capetanaki, Type III intermediate filaments desmin, glial fibrillary acidic protein (GFAP), vimentin, and peripherin. 2017. 9(12): p. a021642.
57. Yadav, R., M. Batra, and R. Chauhan, Role of immunohistochemistry for diagnosis of non-infectious diseases. 2021.
58. Battaglia, R.A., Proteostasis of Glial Intermediate Filaments: Disease Models, Tools, and Mechanisms. 2021, The University of North Carolina at Chapel Hill.
59. Sethi, Y., et al., Broken Heart Syndrome: Evolving Molecular Mechanisms and Principles of Management. 2023. 12(1): p. 125.
60. Mónico, A.M.d.O., Lipoxidación de vimentina y su interacción con zinc. 2020. 61. Topaloğlu, U., H. Sağsöz, and M.E.J.T. Akbalik, Distribution of cytoskeletal proteins in the cat testis during the pre-pubertal and post-pubertal periods. 2023. 197: p. 1-9.
62. Tzioras, M., et al., Invited Review: APOE at the interface of inflammation, neurodegeneration and pathological protein spread in Alzheimer's disease. 2019. 45(4): p. 327-346.
63. Hénaut, L., et al., Cellular and molecular mechanisms associated with ischemic stroke severity in female mice with chronic kidney disease. 2019. 9(1): p. 1-11.
64. Berry, K., et al., Hepatic and renal function impact concentrations of plasma biomarkers of neuropathology. 2022. 14(1): p. e12321.
65. Iqbal, Z., et al., Astrocyte L-Lactate Signaling in the ACC Regulates Visceral Pain Aversive Memory in Rats. 2023. 12(1): p. 26.
66. Shahub, S., et al., A Proof-of-Concept Electrochemical Skin Sensor for Simultaneous Measurement of Glial Fibrillary Acidic Protein (GFAP) and Interleukin-6 (IL-6) for Management of Traumatic Brain Injuries. 2022. 12(12): p. 1095.
67. Kadry, H., et al., A blood–brain barrier overview on structure, function, impairment, and biomarkers of integrity. 2020. 17(1): p. 1-24.
68. Boorman, D.C. and K.A.J.J.o.N.R. Keay, Sex differences in morphine sensitivity are associated with differential glial expression in the brainstem of rats with neuropathic pain. 2022. 100(10): p. 1890-1907.
69. Daly, C.M., et al., Sex differences in response to a high fat, high sucrose diet in both the gut microbiome and hypothalamic astrocytes and microglia. 2022. 25(2): p. 321-335.
70. Trautz, F., et al., Survival-time dependent increase in neuronal IL-6 and astroglial GFAP expression in fatally injured human brain tissue. 2019. 9(1): p. 1-15.
71. Zheng, Z., et al., Albumins as Extracellular Protein Nanoparticles Collaborate with Plasma Ions to Control Biological Osmotic Pressure. 2022: p. 4743-4756.
72. Hladky, S.B., M.A.J.F. Barrand, and B.o.t. CNS, Elimination of substances from the brain parenchyma: efflux via perivascular pathways and via the blood–brain barrier. 2018. 15(1): p. 1-73.
73. Gaudet, A.D. and L.K.J.N. Fonken, Glial cells shape pathology and repair after spinal cord injury. 2018. 15(3): p. 554-577.
74. Zhang, C., et al., Spatiotemporal dynamics of the cellular components involved in glial scar formation following spinal cord injury. 2022. 153: p. 113500.
75. Abdelkareem, R.M., et al., Immunohistochemical expression of nestin as cancer stem cell marker in gliomas. 2019. 5(1): p. 047-051.
76. Bomont, P.J.C.O.i.C.B., The dazzling rise of neurofilaments: Physiological functions and roles as biomarkers. 2021. 68: p. 181-191.
77. Ogorevc, M., et al., Differences in Immunohistochemical and Ultrastructural Features between Podocytes and Parietal Epithelial Cells (PECs) Are Observed in Developing, Healthy Postnatal, and Pathologically Changed Human Kidneys. 2022. 23(14): p. 7501.
78. Agarwal, S., et al., Renal cell markers: lighthouses for managing renal diseases. 2021. 321(6): p. F715-F739.
79. Bornstein, S.R., I. Berger, and C.J.S. Steenblock, Are Nestin-positive cells responsive to stress? 2020. 23(6): p. 662-666.
80. Zoja, C., C. Xinaris, and D.J.F.i.P. Macconi, Diabetic nephropathy: novel molecular mechanisms and therapeutic targets. 2020. 11: p. 586892.
81. Fu, Z. and Y.J.C.M. Yuan, The role of tumor neogenesis pipelines in tumor progression and their therapeutic potential. 2023. 12(2): p. 1558-1571.
82. Mariniello, K., et al., Stem cells, self-renewal, and lineage commitment in the endocrine system. 2019. 10: p. 772.
83. Jaramillo-Rangel, G., et al., Nestin-expressing cells in the lung: the bad and the good parts. 2021. 10(12): p. 3413.
84. Alirezaei, Z., et al., Neurofilament light chain as a biomarker, and correlation with magnetic resonance imaging in diagnosis of CNS-related disorders. 2020. 57(1): p. 469-491.
85. Travica, N., M. Berk, and W.J.C.o.i.p. Marx, Neurofilament light protein as a biomarker in depression and cognitive function. 2022. 35(1): p. 30-37.
86. Garcia-Gancedo, L., et al., Objectively monitoring amyotrophic lateral sclerosis patient symptoms during clinical trials with sensors: observational study. 2019. 7(12): p. e13433.
87. Ferreira-Atuesta, C., et al., The evolution of neurofilament light chain in multiple sclerosis. 2021. 15.
88. Zhao, Y., et al., Neurofilament Light (NF-L) Chain Protein from a Highly Polymerized Structural Component of the Neuronal Cytoskeleton to a Neurodegenerative Disease Biomarker in the Periphery. 2021. 7(2).
89. Akamine, S., et al., Renal function is associated with blood neurofilament light chain level in older adults. 2020. 10(1): p. 1-7.
90. Morgenstern, J., et al., Neuron-specific biomarkers predict hypo-and hyperalgesia in individuals with diabetic peripheral neuropathy. 2021. 64(12): p. 2843-2855.
91. van der Plas, E., et al., Associations between neurofilament light-chain protein, brain structure, and chronic kidney disease. 2022. 91(7): p. 1735-1740.
92. Hsu, Y.-K., et al., Evaluating brain white matter hyperintensity, IQ scores, and plasma neurofilament light chain concentration in early-treated patients with infantile-onset Pompe disease. 2022.
93. Hermann, P., et al., Plasma Lipocalin 2 in Alzheimer’s disease: potential utility in the differential diagnosis and relationship with other biomarkers. 2022. 14: p. 1-12.
94. Marra, C.M., et al., Serum Neurofilament Light in Neurosyphilis: A Pilot Study. 2023. 50(1): p. 42-44.
95. Hou, Y.-C., et al., The role of plasma neurofilament light protein for assessing cognitive impairment in patients with end-stage renal disease. 2021: p. 267.
96. Hawn, S.E., et al., Methylation of the AIM2 gene: An epigenetic mediator of PTSD‐related inflammation and neuropathology plasma biomarkers. 2022. 39(4): p. 323-333.
97. Bavato, F., et al., Altered neuroaxonal integrity in schizophrenia and major depressive disorder assessed with neurofilament light chain in serum. 2021. 140: p. 141-148.
98. de Natale, E.R., et al., How molecular imaging studies can disentangle disease mechanisms in age-related neurodegenerative disorders. 2023: p. 455-492.
99. Zhang, Y., et al., Calcium pyruvate attenuates fat deposition by augmenting fatty acid oxidation and inhibiting glucose oxidation in juvenile large yellow croaker (Larimichthys crocea) consuming a high-fat diet. 2023. 562: p. 738778.
100. Binder, L.I., A. Frankfurter, and L.I.J.T.J.o.c.b. Rebhun, The distribution of tau in the mammalian central nervous system. 1985. 101(4): p. 1371-1378.
101. Kawata, K., et al., Blood biomarkers for brain injury: What are we measuring? 2016. 68: p. 460-473.
102. Johnson, V.E., W. Stewart, and D.H.J.E.n. Smith, Axonal pathology in traumatic brain injury. 2013. 246: p. 35-43.
103. Lee, D.A., et al., Significance of serum neuron-specific enolase in transient global amnesia. 2021. 89: p. 15-19.
104. Roh, H.-T., W.-Y.J.J.o.s. So, and h. science, The effects of aerobic exercise training on oxidant–antioxidant balance, neurotrophic factor levels, and blood–brain barrier function in obese and non-obese men. 2017. 6(4): p. 447-453.
105. Nass, R.D., et al., The role of postictal laboratory blood analyses in the diagnosis and prognosis of seizures. 2017. 47: p. 51-65.
106. Lindblad, C., et al., Influence of blood–brain barrier integrity on brain protein biomarker clearance in severe traumatic brain injury: a longitudinal prospective study. 2020. 37(12): p. 1381-1391.
107. Bailes, J.E., et al., Role of subconcussion in repetitive mild traumatic brain injury: a review. 2013. 119(5): p. 1235-1245.
108. Ayubcha, C., et al., Radiotracers, Positron Emission Tomography Imaging and Traumatic Brain Injury, in Biomarkers in Trauma, Injury and Critical Care. 2022, Springer. p. 1-21.
109. Keating, C.E. and D.K.J.N.o.d. Cullen, Mechanosensation in traumatic brain injury. 2021. 148: p. 105210.
110. Ahmadzadeh, H., D.H. Smith, and V.B.J.B.j. Shenoy, Viscoelasticity of tau proteins leads to strain rate-dependent breaking of microtubules during axonal stretch injury: predictions from a mathematical model. 2014. 106(5): p. 1123-1133.
111. Nowak, M.K., Effects of Subconcussive Head Impacts on Neural Integrity and Function in Attention-Deficit/Hyperactivity Disorder. 2022: Indiana University.
112. Chesser, A.S., S.M. Pritchard, and G.V.J.F.i.n. Johnson, Tau clearance mechanisms and their possible role in the pathogenesis of Alzheimer disease. 2013. 4: p. 122.
113. Tompa, P., et al., On the sequential determinants of calpain cleavage. 2004. 279(20): p. 20775-20785.
114. Zhang, L., R. Sheng, and Z.J.A.b.e.b.S. Qin, The lysosome and neurodegenerative diseases. 2009. 41(6): p. 437-445.
115. Atlante, A., et al., A peptide containing residues 26–44 of tau protein impairs mitochondrial oxidative phosphorylation acting at the level of the adenine nucleotide translocator. 2008. 1777(10): p. 1289-1300.
116. Lynn, K.S., R.J. Peterson, and M.J.B.e.B.A.-B. Koval, Ruffles and spikes: Control of tight junction morphology and permeability by claudins. 2020. 1862(9): p. 183339.
117. Pan, Y.-Y., et al., Structure Composition and Intracellular Transport of Clathrin-Mediated Intestinal Transmembrane Tight Junction Protein. 2022: p. 1-17.
118. Kohlway, A.S., Cause and effect: Hepatitis C virus infection and host innate immune response. 2013: Yale University. 119. Fuladi, S., et al., Computational modeling of claudin structure and function. 2020. 21(3): p. 742.
120. Adil, M.S., S.P. Narayanan, and P.R.J.T.B. Somanath, Cell-cell junctions: structure and regulation in physiology and pathology. 2021. 9(1): p. 1848212.
121. Bhat, A.A., et al., Claudin-1, a double-edged sword in cancer. 2020. 21(2): p. 569.
122. HASELOFF, R.F. and I.E.J.N. BLASIG, HANNELORE BAUER, HANS CHRISTIAN BAUER. 2005: p. 325.
123. Wang, H., X.J.I.J.o.C. Yang, and E. Pathology, The expression patterns of tight junction protein claudin-1,-3, and-4 in human gastric neoplasms and adjacent non-neoplastic tissues. 2015. 8(1): p. 881.
124. Singh, A.B., A. Sharma, and P.J.J.o.o. Dhawan, Claudin family of proteins and cancer: an overview. 2010. 2010.
125. Heijink, I.H., et al., Epithelial cell dysfunction, a major driver of asthma development. 2020. 75(8): p. 1902-1917.
126. Shil, S., Master Of Veterinary Science. 2014, Anand Agricultural University.
127. Yu, A.J.J.o.t.A.S.o.N.J., Claudins and the kidney. 2014. 26(1): p. 11-19.
128. Günzel, D. and A.S.J.P.r. Yu, Claudins and the modulation of tight junction permeability. 2013. 93(2): p. 525-569.
129. Seeger-Nukpezah, T., et al., The hallmarks of cancer: relevance to the pathogenesis of polycystic kidney disease. 2015. 11(9): p. 515-534.
130. Gong, Y. and J.J.P.A.-E.J.o.P. Hou, Claudins in barrier and transport function—the kidney. 2017. 469: p. 105-113.

Most read articles by the same author(s)