| 研究生: |
林羽唯 Lin, Yu-Wei |
|---|---|
| 論文名稱: |
解析急性腎損傷中增生細胞特徵與重組 IL-33對急性腎損傷的保護角色 Characterizing Proliferating Cell Profiles and the Renal Protective Role of Recombinant IL-33 in Acute Kidney Injury |
| 指導教授: |
陳威宇
Chen, Wei-Yu |
| 學位類別: |
碩士 Master |
| 系所名稱: |
醫學院 - 生物化學暨分子生物學研究所 Department of Biochemistry and Molecular Biology |
| 論文出版年: | 2025 |
| 畢業學年度: | 113 |
| 語文別: | 英文 |
| 論文頁數: | 86 |
| 中文關鍵詞: | 急性腎損傷 、增殖細胞 、腎小管 、纖維化 、慢性腎臟病 、介白素-33 |
| 外文關鍵詞: | acute kidney injury, proliferating cells, renal tubules, fibrosis, chronic kidney disease, IL-33 |
| 相關次數: | 點閱:45 下載:0 |
| 分享至: |
| 查詢本校圖書館目錄 查詢臺灣博碩士論文知識加值系統 勘誤回報 |
急性腎損傷(acute kidney injury, AKI)是一種常見且潛在致命的臨床症候群,其後續可能導致不完全的腎小管修復,加劇進展為慢性腎臟病(chronic kidney disease, CKD),並造成全球的高發病率和高死亡率。在AKI後的修復過程中,腎臟內會於急性期出現一群增生細胞,然而目前尚未有系統性的研究解析腎臟中增生細胞組成與時空分布特徵。此外,近年研究指出IL-33的受體ST2表現於多種免疫細胞族群,而第二型先天性淋巴細胞(ILC2)與M2巨噬細胞在腎臟修復中扮演重要角色。過去我們實驗室的研究中亦發現IL-33對於心臟纖維化有保護作用,且通過流式細胞分析後也發現ILC2及巨噬細胞為主要的擴增細胞群。而IL-33作為其主要活化因子,其對於腎臟增生細胞族群的調控機制仍待釐清。本研究以葉酸誘導急性腎損傷(FA-AKI)作為急性腎損傷小鼠模型,並透過BrdU標記追蹤增生細胞。後續結合H&E及Picro-sirius red染色評估組織病理學及纖維化進程。另外以免疫組織染色與定量分析,解析腎臟修復過程中的細胞增生特徵。我們進一步施予重組IL-33(recombinant IL-33)進行治療,探討其是否能促進腎臟組織再生,並透免疫螢光染色分析增生細胞種類,評估IL-33的腎臟保護作用。實驗結果顯示,IL-33治療可顯著提升腎臟功能恢復,減少腎小管損傷,同時也發現IL-33治療則顯著降低全體增生細胞的數量,而其中近端腎小管及間質細胞則為主要的增生細胞,並且也是在IL-33治療後顯著降低的兩種主要細胞族群。本研究揭示IL-33於急性腎損傷後調控腎臟細胞修復及降低腎臟損傷之潛力,並提供一套系統性策略用以解析FA-AKI修復過程中增生細胞的動態分布,有助於發展未來針對腎臟再生的治療策略。
Acute kidney injury (AKI) is a prevalent and clinically significant condition associated with high morbidity and mortality worldwide. A major concern in AKI is the risk of incomplete tubular repair, which can drive the transition to chronic kidney disease (CKD). In the early phase of injury, the kidney responds with a marked increase in proliferating cells, yet the cellular composition and temporal dynamics of these proliferative populations remain only partially understood. Recent studies have identified the IL-33 receptor ST2 on certain immune cell subsets, including type 2 innate lymphoid cells (ILC2s), which are known to play key roles in tissue repair. Our previous study also demonstrated that IL-33 exerts a protective role against cardiac fibrosis, with flow cytometric analysis identifying ILC2s and macrophages as the primary cell populations expanded upon IL-33 stimulation. However, how IL-33 regulates renal proliferative responses remains poorly understood. In this study, we utilized a folic acid–induced acute kidney injury (FA-AKI) mouse model to explore the role of IL-33 in renal repair. Proliferating cells were tracked using BrdU incorporation, and immunofluorescence staining combined with quantitative analysis was used to characterize proliferative profiles during the recovery phase. Recombinant IL-33 was administered therapeutically to evaluate its regenerative capacity and its impact on the composition of proliferating cells. Our results showed that IL-33 treatment significantly improved renal function recovery and attenuated tubular injury. Interestingly, IL-33 also led to a marked reduction in the total number of proliferating cells. Among these, proximal tubular epithelial cells and interstitial cells were identified as the two major proliferative populations most affected by IL-33 administration. In conclusion, this study highlights the role of IL-33 in modulating renal cell proliferation and mitigating fibrosis following AKI. It also provides a systematic approach to profiling proliferative cell populations during FA-AKI recovery, offering insights for the development for future regenerative strategies targeting kidney repair.
Ian V. Murray, M.A.P., Histology, Kidney and Glomerulus. 2023.
Human Osmoregulatory and Excretory Systems - Nephron- The Functional Unit of the Kidney. Vol. 41.11.
Nakhoul, N. and V. Batuman, Role of proximal tubules in the pathogenesis of kidney disease. Contrib Nephrol, 2011. 169: p. 37-50.
Chia, P.Y., A. Teo, and T.W. Yeo, Overview of the Assessment of Endothelial Function in Humans. Front Med (Lausanne), 2020. 7: p. 542567.
Rudman-Melnick, V., et al., Gucy1alpha1 specifically marks kidney, heart, lung and liver fibroblasts. bioRxiv, 2024: p. 594404.
Qu, Z., et al., Tissue-resident macrophages and renal diseases: landscapes and treatment directions. Front Immunol, 2025. 16: p. 1548053.
Murphy, J.M., et al., Tissue-Dependent Adaptations and Functions of Innate Lymphoid Cells. Front Immunol, 2022. 13: p. 836999.
Khwaja, A., KDIGO clinical practice guidelines for acute kidney injury. Nephron Clin Pract, 2012. 120(4): p. c179-84.
Mehta, R.L., et al., Acute Kidney Injury Network: report of an initiative to improve outcomes in acute kidney injury. Crit Care, 2007. 11(2): p. R31.
Basile, D.P., M.D. Anderson, and T.A. Sutton, Pathophysiology of acute kidney injury. Compr Physiol, 2012. 2(2): p. 1303-53.
Gaut, J.P. and H. Liapis, Acute kidney injury pathology and pathophysiology: a retrospective review. Clin Kidney J, 2021. 14(2): p. 526-536.
Hall, A.M. and S. de Seigneux, Metabolic mechanisms of acute proximal tubular injury. Pflugers Arch, 2022. 474(8): p. 813-827.
Perazella, M.A., Drug-induced acute kidney injury: diverse mechanisms of tubular injury. Curr Opin Crit Care, 2019. 25(6): p. 550-557.
Kellum, J.A., et al., Acute kidney injury. Nat Rev Dis Primers, 2021. 7(1): p. 52.
Yao, W., et al., Single Cell RNA Sequencing Identifies a Unique Inflammatory Macrophage Subset as a Druggable Target for Alleviating Acute Kidney Injury. Adv Sci (Weinh), 2022. 9(12): p. e2103675.
Komaru, Y., et al., Acute kidney injury triggers hypoxemia by lung intravascular neutrophil retention that reduces capillary blood flow. J Clin Invest, 2025. 135(10).
Ishani, A., et al., Acute kidney injury increases risk of ESRD among elderly. J Am Soc Nephrol, 2009. 20(1): p. 223-8.
Wang, Z. and C. Zhang, From AKI to CKD: Maladaptive Repair and the Underlying Mechanisms. Int J Mol Sci, 2022. 23(18).
Zhang, T., R.E. Widdop, and S.D. Ricardo, Transition from acute kidney injury to chronic kidney disease: mechanisms, models, and biomarkers. Am J Physiol Renal Physiol, 2024. 327(5): p. F788-F805.
Hsueh, W. and H.H. Rostorfer, Chemically induced renal hypertrophy in the rat. Lab Invest, 1973. 29(5): p. 547-55.
Yan, L.J., Folic acid-induced animal model of kidney disease. Animal Model Exp Med, 2021. 4(4): p. 329-342.
Thorn, C.F., et al., Doxorubicin pathways: pharmacodynamics and adverse effects. Pharmacogenet Genomics, 2011. 21(7): p. 440-6.
Afsar, T., et al., Doxorubicin-induced alterations in kidney functioning, oxidative stress, DNA damage, and renal tissue morphology; Improvement by Acacia hydaspica tannin-rich ethyl acetate fraction. Saudi J Biol Sci, 2020. 27(9): p. 2251-2260.
Eltzschig, H.K. and T. Eckle, Ischemia and reperfusion--from mechanism to translation. Nat Med, 2011. 17(11): p. 1391-401.
Schlingmann, K.P., et al., Germline De Novo Mutations in ATP1A1 Cause Renal Hypomagnesemia, Refractory Seizures, and Intellectual Disability. Am J Hum Genet, 2018. 103(5): p. 808-816.
Hara-Chikuma, M. and A.S. Verkman, Aquaporin-1 facilitates epithelial cell migration in kidney proximal tubule. J Am Soc Nephrol, 2006. 17(1): p. 39-45.
Aggarwal, S., et al., SOX9 switch links regeneration to fibrosis at the single-cell level in mammalian kidneys. Science, 2024. 383(6685): p. eadd6371.
Nie, H., et al., Activated SOX9+ renal epithelial cells promote kidney repair through secreting factors. Cell Prolif, 2023. 56(4): p. e13394.
Song, J., et al., Understanding kidney injury molecule 1: a novel immune factor in kidney pathophysiology. Am J Transl Res, 2019. 11(3): p. 1219-1229.
Zhang, Z. and C.X. Cai, Kidney injury molecule-1 (KIM-1) mediates renal epithelial cell repair via ERK MAPK signaling pathway. Mol Cell Biochem, 2016. 416(1-2): p. 109-16.
Zhang, Z., B.D. Humphreys, and J.V. Bonventre, Shedding of the urinary biomarker kidney injury molecule-1 (KIM-1) is regulated by MAP kinases and juxtamembrane region. J Am Soc Nephrol, 2007. 18(10): p. 2704-14.
Han, W.K., et al., Kidney Injury Molecule-1 (KIM-1): a novel biomarker for human renal proximal tubule injury. Kidney Int, 2002. 62(1): p. 237-44.
Brilland, B., et al., Kidney injury molecule 1 (KIM-1): a potential biomarker of acute kidney injury and tubulointerstitial injury in patients with ANCA-glomerulonephritis. Clin Kidney J, 2023. 16(9): p. 1521-1533.
Schmidt-Ott, K.M., et al., Dual action of neutrophil gelatinase-associated lipocalin. J Am Soc Nephrol, 2007. 18(2): p. 407-13.
Jung, M., et al., Macrophage-derived Lipocalin-2 contributes to ischemic resistance mechanisms by protecting from renal injury. Sci Rep, 2016. 6: p. 21950.
Chen, J.J., et al., Using lipocalin as a prognostic biomarker in acute kidney injury. Expert Rev Mol Diagn, 2021. 21(5): p. 455-464.
Abella, V., et al., The potential of lipocalin-2/NGAL as biomarker for inflammatory and metabolic diseases. Biomarkers, 2015. 20(8): p. 565-71.
Qiu, S., et al., Lipocalin-2 protects against renal ischemia/reperfusion injury in mice through autophagy activation mediated by HIF1alpha and NF-kappab crosstalk. Biomed Pharmacother, 2018. 108: p. 244-253.
Meng, X.M., et al., Innate immune cells in acute and chronic kidney disease. Nat Rev Nephrol, 2025. 21(7): p. 464-482.
Kirita, Y., et al., Cell profiling of mouse acute kidney injury reveals conserved cellular responses to injury. Proc Natl Acad Sci U S A, 2020. 117(27): p. 15874-15883.
Melchinger, I., et al., VCAM-1 mediates proximal tubule-immune cell cross talk in failed tubule recovery during AKI-to-CKD transition. Am J Physiol Renal Physiol, 2024. 327(4): p. F610-F622.
Ferenbach, D.A. and J.V. Bonventre, Mechanisms of maladaptive repair after AKI leading to accelerated kidney ageing and CKD. Nat Rev Nephrol, 2015. 11(5): p. 264-76.
Venkatachalam, M.A., et al., Acute kidney injury: a springboard for progression in chronic kidney disease. Am J Physiol Renal Physiol, 2010. 298(5): p. F1078-94.
Coca, S.G., S. Singanamala, and C.R. Parikh, Chronic kidney disease after acute kidney injury: a systematic review and meta-analysis. Kidney Int, 2012. 81(5): p. 442-8.
Meng, X., J. Jin, and H.Y. Lan, Driving role of macrophages in transition from acute kidney injury to chronic kidney disease. Chin Med J (Engl), 2022. 135(7): p. 757-766.
Balzer, M.S., T. Rohacs, and K. Susztak, How Many Cell Types Are in the Kidney and What Do They Do? Annu Rev Physiol, 2022. 84: p. 507-531.
Li, Z.L., et al., Renal tubular epithelial cells response to injury in acute kidney injury. EBioMedicine, 2024. 107: p. 105294.
Smeets, B., et al., Proximal tubular cells contain a phenotypically distinct, scattered cell population involved in tubular regeneration. J Pathol, 2013. 229(5): p. 645-59.
Cao, J.Y., et al., Exosomal miR-125b-5p deriving from mesenchymal stem cells promotes tubular repair by suppression of p53 in ischemic acute kidney injury. Theranostics, 2021. 11(11): p. 5248-5266.
Berger, K., et al., Origin of regenerating tubular cells after acute kidney injury. Proc Natl Acad Sci U S A, 2014. 111(4): p. 1533-8.
De Chiara, L., et al., Tubular cell polyploidy protects from lethal acute kidney injury but promotes consequent chronic kidney disease. Nat Commun, 2022. 13(1): p. 5805.
Gerhardt, L.M.S., et al., Single-nuclear transcriptomics reveals diversity of proximal tubule cell states in a dynamic response to acute kidney injury. Proc Natl Acad Sci U S A, 2021. 118(27): p. e2026684118.
Zhong, X., et al., Tubular epithelial cells-derived small extracellular vesicle-VEGF-A promotes peritubular capillary repair in ischemic kidney injury. NPJ Regen Med, 2022. 7(1): p. 73.
Zhou, D., et al., Early activation of fibroblasts is required for kidney repair and regeneration after injury. FASEB J, 2019. 33(11): p. 12576-12587.
Tanabe, K., J. Wada, and Y. Sato, Targeting angiogenesis and lymphangiogenesis in kidney disease. Nat Rev Nephrol, 2020. 16(5): p. 289-303.
Humphreys, B.D. and J.V. Bonventre, Mesenchymal stem cells in acute kidney injury. Annu Rev Med, 2008. 59: p. 311-25.
Gui, Y., et al., Fibroblast expression of transmembrane protein smoothened governs microenvironment characteristics after acute kidney injury. J Clin Invest, 2024. 134(13): p. e165836.
Zhou, D., et al., Fibroblast-Specific beta-Catenin Signaling Dictates the Outcome of AKI. J Am Soc Nephrol, 2018. 29(4): p. 1257-1271.
Ye, Y., et al., Pyruvate kinase M2 mediates fibroblast proliferation to promote tubular epithelial cell survival in acute kidney injury. FASEB J, 2021. 35(7): p. e21706.
Lee, K., H.R. Jang, and H. Rabb, Lymphocytes and innate immune cells in acute kidney injury and repair. Nat Rev Nephrol, 2024. 20(12): p. 789-805.
Kurts, C., et al., Kidney immunology from pathophysiology to clinical translation. Nat Rev Immunol, 2025. 25(6): p. 460-476.
do Valle Duraes, F., et al., Immune cell landscaping reveals a protective role for regulatory T cells during kidney injury and fibrosis. JCI Insight, 2020. 5(3): p. e130651.
Goh, C.C., et al., Inducing Ischemia-reperfusion Injury in the Mouse Ear Skin for Intravital Multiphoton Imaging of Immune Responses. J Vis Exp, 2016(118): p. 54956.
Burfeind, K.G., et al., Natural Killer Lymphocytes Mediate Renal Fibrosis Due to Acute Cardiorenal Syndrome. Kidney360, 2024. 5(1): p. 8-21.
Cayrol, C. and J.P. Girard, Interleukin-33 (IL-33): A nuclear cytokine from the IL-1 family. Immunol Rev, 2018. 281(1): p. 154-168.
Chen, W.Y., L.C. Li, and J.L. Yang, Emerging Roles of IL-33/ST2 Axis in Renal Diseases. Int J Mol Sci, 2017. 18(4): p. 783.
Ferhat, M., et al., Endogenous IL-33 Contributes to Kidney Ischemia-Reperfusion Injury as an Alarmin. J Am Soc Nephrol, 2018. 29(4): p. 1272-1288.
Chen, W.Y., et al., Upregulation of Interleukin-33 in obstructive renal injury. Biochem Biophys Res Commun, 2016. 473(4): p. 1026-1032.
Chen, W.Y., et al., IL-33/ST2 axis mediates hyperplasia of intrarenal urothelium in obstructive renal injury. Exp Mol Med, 2018. 50(4): p. 1-11.
Griesenauer, B. and S. Paczesny, The ST2/IL-33 Axis in Immune Cells during Inflammatory Diseases. Front Immunol, 2017. 8: p. 475.
Nagashima, R. and M. Iyoda, The Roles of Kidney-Resident ILC2 in Renal Inflammation and Fibrosis. Front Immunol, 2021. 12: p. 688647.
Sabapathy, V., et al., ST2 + T-Regulatory Cells in Renal Inflammation and Fibrosis after Ischemic Kidney Injury. J Am Soc Nephrol, 2025. 36(1): p. 73-86.
Akcay, A., et al., IL-33 exacerbates acute kidney injury. J Am Soc Nephrol, 2011. 22(11): p. 2057-67.
Park, G.H., et al., Anti-interleukin-33 Reduces Ovalbumin-Induced Nephrotoxicity and Expression of Kidney Injury Molecule-1. Int Neurourol J, 2016. 20(2): p. 114-21.
Cao, Q., et al., Potentiating Tissue-Resident Type 2 Innate Lymphoid Cells by IL-33 to Prevent Renal Ischemia-Reperfusion Injury. J Am Soc Nephrol, 2018. 29(3): p. 961-976.
He, R., et al., IL-33 improves wound healing through enhanced M2 macrophage polarization in diabetic mice. Mol Immunol, 2017. 90: p. 42-49.
Chen, W.Y., et al., Group 2 innate lymphoid cells contribute to IL-33-mediated alleviation of cardiac fibrosis. Theranostics, 2021. 11(6): p. 2594-2611.
Zhang, H.M., et al., ILC2s expanded by exogenous IL-33 regulate CD45+CD11b+F4/80high macrophage polarization to alleviate hepatic ischemia-reperfusion injury. Front Immunol, 2022. 13: p. 869365.
Shao, J. and H. Sheng, Amphiregulin promotes intestinal epithelial regeneration: roles of intestinal subepithelial myofibroblasts. Endocrinology, 2010. 151(8): p. 3728-37.
Poole, J.A., et al., Amphiregulin modulates murine lung recovery and fibroblast function following exposure to agriculture organic dust. Am J Physiol Lung Cell Mol Physiol, 2020. 318(1): p. L180-L191.
Perales-Quintana, M.M., et al., Metabolomic and biochemical characterization of a new model of the transition of acute kidney injury to chronic kidney disease induced by folic acid. PeerJ, 2019. 7: p. e7113.
Aparicio-Trejo, O.E., et al., Chronic impairment of mitochondrial bioenergetics and beta-oxidation promotes experimental AKI-to-CKD transition induced by folic acid. Free Radic Biol Med, 2020. 154: p. 18-32.
Muto, Y., et al., Epigenetic reprogramming driving successful and failed repair in acute kidney injury. Sci Adv, 2024. 10(32): p. eado2849.
Sako, K., et al., Cyclin-dependent kinase 4-related tubular epithelial cell proliferation is regulated by Paired box gene 2 in kidney ischemia-reperfusion injury. Kidney Int, 2022. 102(1): p. 45-57.
Wang, Y., et al., Macrophage-Derived Type 1 IFN, Renal Tubular Epithelial Cell Polyploidization, and AKI-to-CKD Transition. J Am Soc Nephrol, 2025. 36(5): p. 766-780.
Lazzeri, E., et al., Surviving Acute Organ Failure: Cell Polyploidization and Progenitor Proliferation. Trends Mol Med, 2019. 25(5): p. 366-381.
Zhao, Q., et al., FOXQ1, deubiquitinated by USP10, alleviates sepsis-induced acute kidney injury by targeting the CREB5/NF-kappaB signaling axis. Biochim Biophys Acta Mol Basis Dis, 2024. 1870(7): p. 167331.
Chi, Y., et al., microRNA-206 prevents hepatocellular carcinoma growth and metastasis via down-regulating CREB5 and inhibiting the PI3K/AKT signaling pathway. Cell Cycle, 2022. 21(24): p. 2651-2663.
Li, X., et al., FAM3A plays a key role in protecting against tubular cell pyroptosis and acute kidney injury. Redox Biol, 2024. 74: p. 103225.
Babaeenezhad, E., et al., D-Limonene Alleviates Acute Kidney Injury Following Gentamicin Administration in Rats: Role of NF-kappaB Pathway, Mitochondrial Apoptosis, Oxidative Stress, and PCNA. Oxid Med Cell Longev, 2021. 2021: p. 6670007.
Xu, Z., et al., Effect of sirolimus on arteriosclerosis induced by advanced glycation end products via inhibition of the ILK/mTOR pathway in kidney transplantation recipients. Eur J Pharmacol, 2017. 813: p. 1-9.
Galichon, P., et al., Energy depletion by cell proliferation sensitizes the kidney epithelial cells to injury. Am J Physiol Renal Physiol, 2024. 326(3): p. F326-F337.
Wang, Y., et al., ACSL4 deficiency confers protection against ferroptosis-mediated acute kidney injury. Redox Biol, 2022. 51: p. 102262.
Song, M., et al., Alarmin IL-33 orchestrates antitumoral T cell responses to enhance sensitivity to 5-fluorouracil in colorectal cancer. Theranostics, 2023. 13(5): p. 1649-1668.
Jackson-Jones, L.H., et al., IL-33 delivery induces serous cavity macrophage proliferation independent of interleukin-4 receptor alpha. Eur J Immunol, 2016. 46(10): p. 2311-2321.
Kumar, S., et al., Sox9 Activation Highlights a Cellular Pathway of Renal Repair in the Acutely Injured Mammalian Kidney. Cell Rep, 2015. 12(8): p. 1325-38.
Cohen, E.S., et al., Oxidation of the alarmin IL-33 regulates ST2-dependent inflammation. Nat Commun, 2015. 6: p. 8327.
Alt, C., et al., Long-Acting IL-33 Mobilizes High-Quality Hematopoietic Stem and Progenitor Cells More Efficiently Than Granulocyte Colony-Stimulating Factor or AMD3100. Biol Blood Marrow Transplant, 2019. 25(8): p. 1475-1485.
校內:2027-07-31公開