1
|
Tsavlis D, Domvri K, Porpodis K, Papoutsopoulou S, Anestakis D, Tzoumaka A, Meditskou S, Symeonidoy K, Spandou E. Erythropoietin Reduces Inflammation, Oxidative Stress, and Apoptosis in a Rat Model of Bleomycin-Induced Idiopathic Pulmonary Fibrosis. J Pers Med 2024; 14:972. [PMID: 39338226 PMCID: PMC11433300 DOI: 10.3390/jpm14090972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/23/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a lethal interstitial disease with unknown etiology and no effective cure, posing a great health burden to society. Erythropoietin (EPO) has been demonstrated to have protective roles in various tissues such as brain, spinal cord, heart, kidney and lung tissues. In this study, we investigate the specific anti-inflammatory, antioxidant and antiapoptotic effects of erythropoietin on lung tissue in a bleomycin-induced rat model of idiopathic pulmonary fibrosis. METHODS Recombinant human EPO or saline was injected, and the animals were monitored for 14 days after bleomycin instillation. Their hematocrit and serum EPO levels were determined. Histological and immunohistochemical analyses were performed. RESULTS The extent of tissue injury, determined through morphometric analysis, was significantly decreased in size in animals treated with erythropoietin. An immunohistochemical analysis of the expression of cyclooxygenase-2 (COX-2), inducible synthase of nitric oxide (i-NOS), metalloproteinase-9 (MMP-9), erythropoietin receptor (EPO-R), and cytochrome-C (cyt-C) found these enzymes to be decreased in a statistically significant manner in animals treated with erythropoietin when compared to a non-treated group. CONCLUSIONS The reduced expression of COX-2, i-NOS, MMP-9, EPO-R, and i-NOS in the lung tissues of animals treated with EPO indicates the anti-inflammatory, antioxidant and antiapoptotic action of erythropoietin, suggesting its potential therapeutic role in pulmonary fibrosis.
Collapse
Affiliation(s)
- Drosos Tsavlis
- Laboratory of Physiology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.T.); (K.S.); (E.S.)
| | - Kalliopi Domvri
- Laboratory of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.D.); (S.M.)
| | - Konstantinos Porpodis
- Department of Pulmonology, Aristotle University of Thessaloniki, General Hospital G. Papanikolaou, 57010 Thessaloniki, Greece;
| | - Stamatia Papoutsopoulou
- Department of Biochemistry and Biotechnology, Faculty of Life Sciences, University of Thessaly, Mezourlo, 41500 Larissa, Greece;
| | - Doxakis Anestakis
- Department of Pathology & Forensic Sciences, Medical School, University of Cyprus, 1678 Nicosia, Cyprus;
| | - Anna Tzoumaka
- Laboratory of Physiology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.T.); (K.S.); (E.S.)
| | - Soultana Meditskou
- Laboratory of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.D.); (S.M.)
| | - Konstantina Symeonidoy
- Laboratory of Physiology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.T.); (K.S.); (E.S.)
| | - Evangelia Spandou
- Laboratory of Physiology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.T.); (K.S.); (E.S.)
| |
Collapse
|
2
|
Kim T, Surapaneni AL, Schmidt IM, Eadon MT, Kalim S, Srivastava A, Palsson R, Stillman IE, Hodgin JB, Menon R, Otto EA, Coresh J, Grams ME, Waikar SS, Rhee EP. Plasma Proteins Associated with Chronic Histopathologic Lesions on Kidney Biopsy. J Am Soc Nephrol 2024; 35:910-922. [PMID: 38656806 PMCID: PMC11230715 DOI: 10.1681/asn.0000000000000358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/17/2024] [Indexed: 04/26/2024] Open
Abstract
Key Points Proteomic profiling identified 35 blood proteins associated with chronic histopathologic lesions in the kidney. Testican-2 was expressed in the glomerulus, released by the kidney into circulation, and inversely associated with glomerulosclerosis severity. NELL1 was expressed in tubular epithelial cells, released by the kidney into circulation, and inversely associated with interstitial fibrosis and tubular atrophy severity. Background The severity of chronic histopathologic lesions on kidney biopsy is independently associated with higher risk of progressive CKD. Because kidney biopsies are invasive, identification of blood markers that report on underlying kidney histopathology has the potential to enhance CKD care. Methods We examined the association between 6592 plasma protein levels measured by aptamers and the severity of interstitial fibrosis and tubular atrophy (IFTA), glomerulosclerosis, arteriolar sclerosis, and arterial sclerosis among 434 participants of the Boston Kidney Biopsy Cohort. For proteins significantly associated with at least one histologic lesion, we assessed renal arteriovenous protein gradients among 21 individuals who had undergone invasive catheterization and assessed the expression of the cognate gene among 47 individuals with single-cell RNA sequencing data in the Kidney Precision Medicine Project. Results In models adjusted for eGFR, proteinuria, and demographic factors, we identified 35 proteins associated with one or more chronic histologic lesions, including 20 specific for IFTA, eight specific for glomerulosclerosis, and one specific for arteriolar sclerosis. In general, higher levels of these proteins were associated with more severe histologic score and lower eGFR. Exceptions included testican-2 and NELL1, which were associated with less glomerulosclerosis and IFTA, respectively, and higher eGFR; notably, both of these proteins demonstrated significantly higher levels from artery to renal vein, demonstrating net kidney release. In the Kidney Precision Medicine Project, 13 of the 35 protein hits had cognate gene expression enriched in one or more cell types in the kidney, including podocyte expression of select glomerulosclerosis markers (including testican-2) and tubular expression of several IFTA markers (including NELL1). Conclusions Proteomic analysis identified circulating proteins associated with chronic histopathologic lesions, some of which had concordant site-specific expression within the kidney.
Collapse
Affiliation(s)
- Taesoo Kim
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Aditya L. Surapaneni
- Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Insa M. Schmidt
- Section of Nephrology, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts
| | - Michael T. Eadon
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Sahir Kalim
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Anand Srivastava
- Division of Nephrology, University of Illinois Chicago, Chicago, Illinois
| | - Ragnar Palsson
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Isaac E. Stillman
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jeffrey B. Hodgin
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Rajasree Menon
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Edgar A. Otto
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Josef Coresh
- Departments of Population Health and Medicine, New York University Grossman School of Medicine, New York, New York
| | - Morgan E. Grams
- Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Sushrut S. Waikar
- Section of Nephrology, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, Massachusetts
| | - Eugene P. Rhee
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
3
|
Yasuoka Y, Izumi Y, Fukuyama T, Oshima T, Yamazaki T, Uematsu T, Kobayashi N, Nanami M, Shimada Y, Nagaba Y, Mukoyama M, Sands JM, Takahashi N, Kawahara K, Nonoguchi H. Tubular Endogenous Erythropoietin Protects Renal Function against Ischemic Reperfusion Injury. Int J Mol Sci 2024; 25:1223. [PMID: 38279224 PMCID: PMC10816907 DOI: 10.3390/ijms25021223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 01/28/2024] Open
Abstract
Many large-scale studies show that exogenous erythropoietin, erythropoiesis-stimulating agents, lack any renoprotective effects. We investigated the effects of endogenous erythropoietin on renal function in kidney ischemic reperfusion injury (IRI) using the prolyl hydroxylase domain (PHD) inhibitor, Roxadustat (ROX). Four h of hypoxia (7% O2) and 4 h treatment by ROX prior to IRI did not improve renal function. In contrast, 24-72 h pretreatment by ROX significantly improved the decline of renal function caused by IRI. Hypoxia and 4 h ROX increased interstitial cells-derived Epo production by 75- and 6-fold, respectively, before IRI, and worked similarly to exogenous Epo. ROX treatment for 24-72 h increased Epo production during IRI by 9-fold. Immunohistochemistry revealed that 24 h ROX treatment induced Epo production in proximal and distal tubules and worked similarly to endogenous Epo. Our data show that tubular endogenous Epo production induced by 24-72 h ROX treatment results in renoprotection but peritubular exogenous Epo production by interstitial cells induced by hypoxia and 4 h ROX treatment did not. Stimulation of tubular, but not peritubular, Epo production may link to renoprotection.
Collapse
Affiliation(s)
- Yukiko Yasuoka
- Department of Physiology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0374, Kanagawa, Japan; (Y.Y.); (T.O.); (K.K.)
| | - Yuichiro Izumi
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Kumamoto, Japan; (Y.I.); (M.M.)
| | - Takashi Fukuyama
- Division of Biomedical Research, Kitasato University Medical Center, 6-100 Arai, Kitamoto 364-8501, Saitama, Japan; (T.F.); (T.Y.); (T.U.); (N.K.)
| | - Tomomi Oshima
- Department of Physiology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0374, Kanagawa, Japan; (Y.Y.); (T.O.); (K.K.)
| | - Taiga Yamazaki
- Division of Biomedical Research, Kitasato University Medical Center, 6-100 Arai, Kitamoto 364-8501, Saitama, Japan; (T.F.); (T.Y.); (T.U.); (N.K.)
| | - Takayuki Uematsu
- Division of Biomedical Research, Kitasato University Medical Center, 6-100 Arai, Kitamoto 364-8501, Saitama, Japan; (T.F.); (T.Y.); (T.U.); (N.K.)
| | - Noritada Kobayashi
- Division of Biomedical Research, Kitasato University Medical Center, 6-100 Arai, Kitamoto 364-8501, Saitama, Japan; (T.F.); (T.Y.); (T.U.); (N.K.)
| | - Masayoshi Nanami
- Division of Kidney and Dialysis, Department of Internal Medicine, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya 663-8501, Hyogo, Japan;
| | - Yoshitaka Shimada
- Division of Internal Medicine, Kitasato University Medical Center, 6-100 Arai, Kitamoto 364-8501, Saitama, Japan; (Y.S.); (Y.N.)
| | - Yasushi Nagaba
- Division of Internal Medicine, Kitasato University Medical Center, 6-100 Arai, Kitamoto 364-8501, Saitama, Japan; (Y.S.); (Y.N.)
| | - Masashi Mukoyama
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Kumamoto, Japan; (Y.I.); (M.M.)
| | - Jeff M. Sands
- Renal Division, Department of Medicine, Emory University School of Medicine, 1639 Pierce Drive, WMB Room 3313, Atlanta, GA 30322, USA;
| | - Noriko Takahashi
- Department of Physiology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0374, Kanagawa, Japan; (Y.Y.); (T.O.); (K.K.)
| | - Katsumasa Kawahara
- Department of Physiology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0374, Kanagawa, Japan; (Y.Y.); (T.O.); (K.K.)
| | - Hiroshi Nonoguchi
- Division of Internal Medicine, Kitasato University Medical Center, 6-100 Arai, Kitamoto 364-8501, Saitama, Japan; (Y.S.); (Y.N.)
| |
Collapse
|
4
|
Tanaka S, Portilla D, Okusa MD. Role of perivascular cells in kidney homeostasis, inflammation, repair and fibrosis. Nat Rev Nephrol 2023; 19:721-732. [PMID: 37608184 DOI: 10.1038/s41581-023-00752-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2023] [Indexed: 08/24/2023]
Abstract
Perivascular niches in the kidney comprise heterogeneous cell populations, including pericytes and fibroblasts, with distinct functions. These perivascular cells have crucial roles in preserving kidney homeostasis as they maintain microvascular networks by stabilizing the vasculature and regulating capillary constriction. A subset of kidney perivascular cells can also produce and secrete erythropoietin; this ability can be enhanced with hypoxia-inducible factor-prolyl hydroxylase inhibitors, which are used to treat anaemia in chronic kidney disease. In the pathophysiological state, kidney perivascular cells contribute to the progression of kidney fibrosis, partly via transdifferentiation into myofibroblasts. Moreover, perivascular cells are now recognized as major innate immune sentinels in the kidney that produce pro-inflammatory cytokines and chemokines following injury. These mediators promote immune cell infiltration, leading to persistent inflammation and progression of kidney fibrosis. The crosstalk between perivascular cells and tubular epithelial, immune and endothelial cells is therefore a key process in physiological and pathophysiological states. Here, we examine the multiple roles of kidney perivascular cells in health and disease, focusing on the latest advances in this field of research.
Collapse
Affiliation(s)
- Shinji Tanaka
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| | - Didier Portilla
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, VA, USA
| | - Mark D Okusa
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
5
|
Wu Y, Huang L, Sai W, Chen F, Liu Y, Han C, Barker JM, Zwaini ZD, Lowe MP, Brunskill NJ, Yang B. HBSP improves kidney ischemia-reperfusion injury and promotes repair in properdin deficient mice via enhancing phagocytosis of tubular epithelial cells. Front Immunol 2023; 14:1183768. [PMID: 37207230 PMCID: PMC10188997 DOI: 10.3389/fimmu.2023.1183768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 04/13/2023] [Indexed: 05/21/2023] Open
Abstract
Phagocytosis plays vital roles in injury and repair, while its regulation by properdin and innate repair receptor, a heterodimer receptor of erythropoietin receptor (EPOR)/β common receptor (βcR), in renal ischaemia-reperfusion (IR) remains unclear. Properdin, a pattern recognition molecule, facilitates phagocytosis by opsonizing damaged cells. Our previous study showed that the phagocytic function of tubular epithelial cells isolated from properdin knockout (PKO) mouse kidneys was compromised, with upregulated EPOR in IR kidneys that was further raised by PKO at repair phase. Here, helix B surface peptide (HBSP), derived from EPO only recognizing EPOR/βcR, ameliorated IR-induced functional and structural damage in both PKO and wild-type (WT) mice. In particular, HBSP treatment led to less cell apoptosis and F4/80+ macrophage infiltration in the interstitium of PKO IR kidneys compared to the WT control. In addition, the expression of EPOR/βcR was increased by IR in WT kidneys, and furthered increased in IR PKO kidneys, but greatly reduced by HBSP in the IR kidneys of PKO mice. HBSP also increased PCNA expression in IR kidneys of both genotypes. Moreover, iridium-labelled HBSP (HBSP-Ir) was localized mainly in the tubular epithelia after 17-h renal IR in WT mice. HBSP-Ir also anchored to mouse kidney epithelial (TCMK-1) cells treated by H2O2. Both EPOR and EPOR/βcR were significantly increased by H2O2 treatment, while further increased EPOR was showed in cells transfected with small interfering RNA (siRNA) targeting properdin, but a lower level of EPOR was seen in EPOR siRNA and HBSP-treated cells. The number of early apoptotic cells was increased by EPOR siRNA in H2O2-treated TCMK-1, but markedly reversed by HBSP. The phagocytic function of TCMK-1 cells assessed by uptake fluorescence-labelled E.coli was enhanced by HBSP dose-dependently. Our data demonstrate for the first time that HBSP improves the phagocytic function of tubular epithelial cells and kidney repair post IR injury, via upregulated EPOR/βcR triggered by both IR and properdin deficiency.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Department of Pathology, Medical School of Nantong University, Nantong, China
- Department of Cardiovascular Sciences, College of Life Sciences, University of Leicester, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Lili Huang
- Nantong-Leicester Joint Institute of Kidney Science, Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Weili Sai
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Fei Chen
- Nantong-Leicester Joint Institute of Kidney Science, Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yu Liu
- Nantong-Leicester Joint Institute of Kidney Science, Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Cheng Han
- Nantong-Leicester Joint Institute of Kidney Science, Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Joanna M. Barker
- School of Chemistry, University of Leicester, Leicester, United Kingdom
| | - Zinah D. Zwaini
- Department of Respiratory Sciences, College of Life Sciences, University of Leicester, Leicester, United Kingdom
| | - Mark P. Lowe
- School of Chemistry, University of Leicester, Leicester, United Kingdom
| | - Nigel J. Brunskill
- Department of Cardiovascular Sciences, College of Life Sciences, University of Leicester, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
- Nantong-Leicester Joint Institute of Kidney Science, Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Bin Yang
- Department of Cardiovascular Sciences, College of Life Sciences, University of Leicester, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
- Nantong-Leicester Joint Institute of Kidney Science, Nephrology, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Bin Yang,
| |
Collapse
|
6
|
Shi M, Maique JO, Cleaver O, Moe OW, Hu MC. VEGFR2 insufficiency enhances phosphotoxicity and undermines Klotho's protection against peritubular capillary rarefaction and kidney fibrosis. Am J Physiol Renal Physiol 2023; 324:F106-F123. [PMID: 36395384 PMCID: PMC9799155 DOI: 10.1152/ajprenal.00149.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 10/17/2022] [Accepted: 11/03/2022] [Indexed: 11/18/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) and its cognate receptor (VEGFR2) system are crucial for cell functions associated with angiogenesis and vasculogenesis. Klotho contributes to vascular health maintenance in the kidney and other organs in mammals, but it is unknown whether renoprotection by Klotho is dependent on VEGF/VEGFR2 signaling. We used heterozygous VEGFR2-haploinsufficient (VEGFR2+/-) mice resulting from heterozygous knockin of green fluorescent protein in the locus of fetal liver kinase 1 encoding VEGFR2 to test the interplay of Klotho, phosphate, and VEGFR2 in kidney function, the vasculature, and fibrosis. VEGFR2+/- mice displayed downregulated VEGF/VEGFR2 signaling in the kidney, lower density of peritubular capillaries, and accelerated kidney fibrosis, all of which were also found in the homozygous Klotho hypomorphic mice. High dietary phosphate induced higher plasma phosphate, greater peritubular capillary rarefaction, and more kidney fibrosis in VEGFR2+/- mice compared with wild-type mice. Genetic overexpression of Klotho significantly attenuated the elevated plasma phosphate, kidney dysfunction, peritubular capillary rarefaction, and kidney fibrosis induced by a high-phosphate diet in wild-type mice but only modestly ameliorated these changes in the VEGFR2+/- background. In cultured endothelial cells, VEGFR2 inhibition reduced free VEGFR2 but enhanced its costaining of an endothelial marker (CD31) and exacerbated phosphotoxicity. Klotho protein maintained VEGFR2 expression and attenuated high phosphate-induced cell injury, which was reduced by VEGFR2 inhibition. In conclusion, normal VEGFR2 function is required for vascular integrity and for Klotho to exert vascular protective and antifibrotic actions in the kidney partially through the regulation of VEGFR2 function.NEW & NOTEWORTHY This research paper studied the interplay of vascular endothelial growth factor receptor type 2 (VEGFR2), high dietary phosphate, and Klotho, an antiaging protein, in peritubular structure and kidney fibrosis. Klotho protein was shown to maintain VEGFR2 expression in the kidney and reduce high phosphate-induced cell injury. However, Klotho cytoprotection was attenuated by VEGFR2 inhibition. Thus, normal VEGFR2 function is required for vascular integrity and Klotho to exert vascular protective and antifibrotic actions in the kidney.
Collapse
Affiliation(s)
- Mingjun Shi
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, Texas
| | - Jenny Omega Maique
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, Texas
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, Texas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, Texas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
7
|
Tanemoto F, Nangaku M, Mimura I. Epigenetic memory contributing to the pathogenesis of AKI-to-CKD transition. Front Mol Biosci 2022; 9:1003227. [PMID: 36213117 PMCID: PMC9532834 DOI: 10.3389/fmolb.2022.1003227] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/24/2022] [Indexed: 11/18/2022] Open
Abstract
Epigenetic memory, which refers to the ability of cells to retain and transmit epigenetic marks to their daughter cells, maintains unique gene expression patterns. Establishing programmed epigenetic memory at each stage of development is required for cell differentiation. Moreover, accumulating evidence shows that epigenetic memory acquired in response to environmental stimuli may be associated with diverse diseases. In the field of kidney diseases, the “memory” of acute kidney injury (AKI) leads to progression to chronic kidney disease (CKD); epidemiological studies show that patients who recover from AKI are at high risk of developing CKD. The underlying pathological processes include nephron loss, maladaptive epithelial repair, inflammation, and endothelial injury with vascular rarefaction. Further, epigenetic alterations may contribute as well to the pathophysiology of this AKI-to-CKD transition. Epigenetic changes induced by AKI, which can be recorded in cells, exert long-term effects as epigenetic memory. Considering the latest findings on the molecular basis of epigenetic memory and the pathophysiology of AKI-to-CKD transition, we propose here that epigenetic memory contributing to AKI-to-CKD transition can be classified according to the presence or absence of persistent changes in the associated regulation of gene expression, which we designate “driving” memory and “priming” memory, respectively. “Driving” memory, which persistently alters the regulation of gene expression, may contribute to disease progression by activating fibrogenic genes or inhibiting renoprotective genes. This process may be involved in generating the proinflammatory and profibrotic phenotypes of maladaptively repaired tubular cells after kidney injury. “Priming” memory is stored in seemingly successfully repaired tubular cells in the absence of detectable persistent phenotypic changes, which may enhance a subsequent transcriptional response to the second stimulus. This type of memory may contribute to AKI-to-CKD transition through the cumulative effects of enhanced expression of profibrotic genes required for wound repair after recurrent AKI. Further understanding of epigenetic memory will identify therapeutic targets of future epigenetic intervention to prevent AKI-to-CKD transition.
Collapse
|
8
|
Dai R, Zhang L, Jin H, Wang D, Cheng M, Sang T, Peng C, Li Y, Wang Y. Autophagy in renal fibrosis: Protection or promotion? Front Pharmacol 2022; 13:963920. [PMID: 36105212 PMCID: PMC9465674 DOI: 10.3389/fphar.2022.963920] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Autophagy is a process that degrades endogenous cellular protein aggregates and damaged organelles via the lysosomal pathway to maintain cellular homeostasis and energy production. Baseline autophagy in the kidney, which serves as a quality control system, is essential for cellular metabolism and organelle homeostasis. Renal fibrosis is the ultimate pathological manifestation of progressive chronic kidney disease. In several experimental models of renal fibrosis, different time points, stimulus intensities, factors, and molecular mechanisms mediating the upregulation or downregulation of autophagy may have different effects on renal fibrosis. Autophagy occurring in a single lesion may also exert several distinct biological effects on renal fibrosis. Thus, whether autophagy prevents or facilitates renal fibrosis remains a complex and challenging question. This review explores the different effects of the dual regulatory function of autophagy on renal fibrosis in different renal fibrosis models, providing ideas for future work in related basic and clinical research.
Collapse
Affiliation(s)
- Rong Dai
- Department of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Lei Zhang
- Department of Nephrology, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Hua Jin
- Department of Nephrology, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Dong Wang
- Department of Nephrology, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Meng Cheng
- Department of Nephrology, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Tian Sang
- Graduate School, Anhui University of Chinese Medicine, Hefei, China
| | - Chuyi Peng
- Graduate School, Anhui University of Chinese Medicine, Hefei, China
| | - Yue Li
- Blood Purification Center, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Yiping Wang
- Department of Nephrology, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- *Correspondence: Yiping Wang,
| |
Collapse
|
9
|
Yi X, Yan W, Guo T, Liu N, Wang Z, Shang J, Wei X, Cui X, Sun Y, Ren S, Chen L. Erythropoietin Mitigates Diabetic Nephropathy by Restoring PINK1/Parkin-Mediated Mitophagy. Front Pharmacol 2022; 13:883057. [PMID: 35656290 PMCID: PMC9152250 DOI: 10.3389/fphar.2022.883057] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetic nephropathy (DN), one of the most detrimental microvascular complications of diabetes, is the leading cause of end-stage renal disease. The pathogenesis of DN is complicated, including hemodynamic changes, inflammatory response, oxidative stress, among others. Recently, many studies have demonstrated that mitophagy, especially PINK1/Parkin-mediated mitophagy, plays an important role in the pathogenesis of DN. Erythropoietin (EPO), a glycoprotein hormone mainly secreted by the kidney, regulates the production of erythrocytes. This research intends to explore the beneficial effects of EPO on DN and investigate related mechanisms. In in vitro experiments, we found that EPO promoted autophagic flux and alleviated mitochondrial dysfunction in terms of mitochondrial fragmentation, elevated mitochondrial ROS as well as the loss of mitochondrial potential, and lowered the apoptosis level in high-glucose-treated mesangial cells. Moreover, EPO increased protein expressions of PINK1 and Parkin, enhanced the co-localization of LC3 with mitochondria, Parkin with mitochondria as well as LC3 with Parkin, and increased the number of GFP-LC3 puncta, resulting in increased level of PINK1/Parkin-mediated mitophagy in mesangial cells. The knockdown of PINK1 abrogated the effect of EPO on mitophagy. In addition, in vivo experiments demonstrated that EPO attenuated renal injury, reduced oxidative stress, and promoted expressions of genes related to PINK1/Parkin-mediated mitophagy in the kidneys of DN mice. In summary, these results suggest that PINK1/Parkin-mediated mitophagy is involved in the development of DN and EPO mitigates DN by restoring PINK1/Parkin-mediated mitophagy.
Collapse
Affiliation(s)
- Xinyao Yi
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Wenhui Yan
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Tingli Guo
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Na Liu
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Zhuanzhuan Wang
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Jia Shang
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Xiaotong Wei
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Xin Cui
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Yuzhuo Sun
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Shuting Ren
- Department of Phathology, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Lina Chen
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
10
|
Shi M, Maique J, Shepard S, Li P, Seli O, Moe OW, Chang Hu M. In vivo evidence for therapeutic applications of beclin 1 to promote recovery and inhibit fibrosis after acute kidney injury. Kidney Int 2022; 101:63-78. [PMID: 34736972 PMCID: PMC8741729 DOI: 10.1016/j.kint.2021.09.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 09/09/2021] [Accepted: 09/16/2021] [Indexed: 01/18/2023]
Abstract
Autophagy regulator beclin 1 activity determines the severity of kidney damage induced by ischemia reperfusion injury, but its role in kidney recovery and fibrosis are unknown and its therapeutic potentials have not been tested. Here, we explored beclin 1 effects on kidney fibrosis in three models of acute kidney injury (AKI)-ischemia reperfusion injury, cisplatin kidney toxicity, and unilateral ureteric obstruction in mouse strains with three levels of beclin 1 function: normal (wild type), low (heterozygous global deletion of beclin 1, Becn1+/-), and high beclin 1 activity (knockin gain-of-function mutant Becn1, Becn1FA). Fourteen days after AKI induction, heterozygous mice had more, but knockin mice had less kidney fibrosis than wild-type mice did. One day after ischemia reperfusion injury, heterozygous pan-kidney tubular Becn1 null mice had more severe kidney damage than homozygous distal tubular Becn1 null mice did, which was similar to the wild-type mice, implying that proximal tubular beclin 1 protects the kidney against ischemia reperfusion injury. By 14 days, both pan-kidney heterozygous Becn1 null and distal tubular homozygous Becn1 null mice had poorer kidney recovery than wild-type mice did. Injection of beclin 1 peptides increased cell proliferation in kidney tubules in normal mice. Beclin 1 peptides injection either before or after (2-5 days) ischemia reperfusion injury protected the kidney from injury and suppressed kidney fibrosis. Thus, both endogenous beclin 1 protein expression in kidney tubules and exogenous beclin 1 peptides are kidney protective via attenuation of acute kidney damage, promotion of cell proliferation, and inhibition of kidney fibrosis, consequently improving kidney recovery post-AKI. Hence, exogenous beclin 1 peptide may be a potential new therapy for AKI.
Collapse
Affiliation(s)
- Mingjun Shi
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research
| | - Jenny Maique
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research
| | - Sierra Shepard
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research
| | - Peng Li
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research
| | - Olivia Seli
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research
| | - Orson W. Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA,Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA,Address for reprint request and other correspondence: Ming Chang Hu, MD, PhD, Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390 USA, or Orson W. Moe, MD, Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390 USA,
| | - Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA,Address for reprint request and other correspondence: Ming Chang Hu, MD, PhD, Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390 USA, or Orson W. Moe, MD, Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390 USA,
| |
Collapse
|
11
|
Chancharoenthana W, Udompronpitak K, Manochantr Y, Kantagowit P, Kaewkanha P, Issara-Amphorn J, Leelahavanichkul A. Repurposing of High-Dose Erythropoietin as a Potential Drug Attenuates Sepsis in Preconditioning Renal Injury. Cells 2021; 10:3133. [PMID: 34831360 PMCID: PMC8617638 DOI: 10.3390/cells10113133] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/07/2021] [Accepted: 11/09/2021] [Indexed: 12/29/2022] Open
Abstract
Due to (i) the uremia-enhanced sepsis severity, (ii) the high prevalence of sepsis with pre-existing renal injury and (iii) the non-erythropoiesis immunomodulation of erythropoietin (EPO), EPO was tested in sepsis with pre-existing renal injury models with the retrospective exploration in patients. Then, EPO was subcutaneously administered in mice with (i) cecal ligation and puncture (CLP) after renal injury including 5/6 nephrectomy (5/6Nx-CLP) and bilateral nephrectomy (BiNx-CLP) or sham surgery (sham-CLP) and (ii) lipopolysaccharide (LPS) injection, along with testing in macrophages. In patients, the data of EPO administration and the disease characteristics in patients with sepsis-induced acute kidney injury (sepsis-AKI) were evaluated. As such, increased endogenous EPO was demonstrated in all sepsis models, including BiNx-CLP despite the reduced liver erythropoietin receptor (EPOR), using Western blot analysis and gene expression, in liver (partly through hepatocyte apoptosis). A high-dose EPO, but not a low-dose, attenuated sepsis in mouse models as determined by mortality and serum inflammatory cytokines. Furthermore, EPO attenuated inflammatory responses in LPS-activated macrophages as determined by supernatant cytokines and the expression of several inflammatory genes (iNOS, IL-1β, STAT3 and NFκB). In parallel, patients with sepsis-AKI who were treated with the high-dose EPO showed favorable outcomes, particularly the 29-day mortality rate. In conclusion, high-dose EPO attenuated sepsis with preconditioning renal injury in mice possibly through the macrophage anti-inflammatory effect, which might be beneficial in some patients.
Collapse
Affiliation(s)
- Wiwat Chancharoenthana
- Tropical Nephrology Research Unit, Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand;
| | - Kanyarat Udompronpitak
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (K.U.); (Y.M.); (P.K.); (P.K.); (J.I.-A.)
| | - Yolradee Manochantr
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (K.U.); (Y.M.); (P.K.); (P.K.); (J.I.-A.)
| | - Piyawat Kantagowit
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (K.U.); (Y.M.); (P.K.); (P.K.); (J.I.-A.)
| | - Ponthakorn Kaewkanha
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (K.U.); (Y.M.); (P.K.); (P.K.); (J.I.-A.)
| | - Jiraporn Issara-Amphorn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (K.U.); (Y.M.); (P.K.); (P.K.); (J.I.-A.)
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (K.U.); (Y.M.); (P.K.); (P.K.); (J.I.-A.)
- Translational Research in Inflammation and Immunology Research Unit (TRITU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
12
|
Zhang Y, Wu Y, Wang W, Liu F, Zhang Y, Yang C, Liu A, Wu J, Zhu T, Nicholson ML, Fan Y, Yang B. Long-Term Protection of CHBP Against Combinational Renal Injury Induced by Both Ischemia-Reperfusion and Cyclosporine A in Mice. Front Immunol 2021; 12:697751. [PMID: 34381450 PMCID: PMC8350137 DOI: 10.3389/fimmu.2021.697751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/25/2021] [Indexed: 11/13/2022] Open
Abstract
Renal ischemia–reperfusion (IR) injury and cyclosporine A (CsA) nephrotoxicity affect allograft function and survival. The prolonged effects and underlying mechanisms of erythropoietin derived cyclic helix B peptide (CHBP) and/or caspase-3 small interfering RNA (CASP-3siRNA) were investigated in mouse kidneys, as well as kidney epithelial cells (TCMK-1), subjected to transplant-related injuries. Bilateral renal pedicles were clamped for 30 min followed by reperfusion for 2 and 8 weeks, with/without 35 mg/kg CsA gavage daily and/or 24 nmol/kg CHBP intraperitoneal injection every 3 days. The ratio of urinary albumin to creatinine was raised by IR injury, further increased by CsA and lowered by CHBP at 2, 4, 6 and 8 weeks, whereas the level of SCr was not significantly affected. Similar change trends were revealed in tubulointerstitial damage and fibrosis, HMGB1 and active CASP-3 protein. Increased apoptotic cells in IR kidneys were decreased by CsA and CHBP at 2 and/or 8 weeks. p70 S6 kinase and mTOR were reduced by CsA with/without CHBP at 2 weeks, so were S6 ribosomal protein and GSK-3β at 8 weeks, with reduced CASP-3 at both time points. CASP-3 was further decreased by CHBP in IR or IR + CsA kidneys at 2 or 8 weeks. Furthermore, in TCMK-1 cells CsA induced apoptosis was decreased by CHBP and/or CASP-3siRNA treatment. Taken together, CHBP predominantly protects kidneys against IR injury at 2 weeks and/or CsA nephrotoxicity at 8 weeks, with different underlying mechanisms. Urinary albumin/creatinine is a good biomarker in monitoring the progression of transplant-related injuries. CsA divergently affects apoptosis in kidneys and cultured kidney epithelial cells, in which CHBP and/or CASP-3siRNA reduces inflammation and apoptosis.
Collapse
Affiliation(s)
- Yufang Zhang
- Renal Group, Basic Medical Research Centre, Medical College of Nantong University, Nantong, China
| | - Yuanyuan Wu
- Renal Group, Basic Medical Research Centre, Medical College of Nantong University, Nantong, China.,Nantong-Leicester Joint Institute of Kidney Science, Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Wei Wang
- Nantong-Leicester Joint Institute of Kidney Science, Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Feng Liu
- Nantong-Leicester Joint Institute of Kidney Science, Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yiwen Zhang
- Nantong-Leicester Joint Institute of Kidney Science, Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Cheng Yang
- Department of Urology, Zhongshan Hospital, Shanghai Key Laboratory of Organ Transplantation, Fudan University, Shanghai, China
| | - Aifen Liu
- Renal Group, Basic Medical Research Centre, Medical College of Nantong University, Nantong, China
| | - Jing Wu
- Nantong-Leicester Joint Institute of Kidney Science, Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Tongyu Zhu
- Department of Urology, Zhongshan Hospital, Shanghai Key Laboratory of Organ Transplantation, Fudan University, Shanghai, China
| | - Michael L Nicholson
- Department of Cardiovascular Sciences, University of Leicester, University Hospitals of Leicester, Leicester, United Kingdom.,Department of Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Yaping Fan
- Nantong-Leicester Joint Institute of Kidney Science, Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Bin Yang
- Renal Group, Basic Medical Research Centre, Medical College of Nantong University, Nantong, China.,Nantong-Leicester Joint Institute of Kidney Science, Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China.,Department of Cardiovascular Sciences, University of Leicester, University Hospitals of Leicester, Leicester, United Kingdom
| |
Collapse
|
13
|
Wu Y, Yang B. Erythropoietin Receptor/β Common Receptor: A Shining Light on Acute Kidney Injury Induced by Ischemia-Reperfusion. Front Immunol 2021; 12:697796. [PMID: 34276689 PMCID: PMC8278521 DOI: 10.3389/fimmu.2021.697796] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 05/17/2021] [Indexed: 12/29/2022] Open
Abstract
Acute kidney injury (AKI) is a health problem worldwide, but there is a lack of early diagnostic biomarkers and target-specific treatments. Ischemia-reperfusion (IR), a major cause of AKI, not only induces kidney injury, but also stimulates the self-defense system including innate immune responses to limit injury. One of these responses is the production of erythropoietin (EPO) by adjacent normal tissue, which is simultaneously triggered, but behind the action of its receptors, either by the homodimer EPO receptor (EPOR)2 mainly involved in erythropoiesis or the heterodimer EPOR/β common receptor (EPOR/βcR) which has a broad range of biological protections. EPOR/βcR is expressed in several cell types including tubular epithelial cells at low levels or absent in normal kidneys, but is swiftly upregulated by hypoxia and inflammation and also translocated to cellular membrane post IR. EPOR/βcR mediates anti-apoptosis, anti-inflammation, pro-regeneration, and remodeling via the PI3K/Akt, STAT3, and MAPK signaling pathways in AKI. However, the precise roles of EPOR/βcR in the pathogenesis and progression of AKI have not been well defined, and its potential as an earlier biomarker for AKI diagnosis and monitoring repair or chronic progression requires further investigation. Here, we review biological functions and mechanistic signaling pathways of EPOR/βcR in AKI, and discuss its potential clinical applications as a biomarker for effective diagnosis and predicting prognosis, as well as directing cell target drug delivery.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Basic Medical Research Centre, Medical School, Nantong University, Nantong, China.,Nantong-Leicester Joint Institute of Kidney Science, Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Bin Yang
- Nantong-Leicester Joint Institute of Kidney Science, Nephrology, Affiliated Hospital of Nantong University, Nantong, China.,Department of Cardiovascular Sciences, College of Life Sciences, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
14
|
Abstract
Renal epithelial cells show remarkable regenerative capacity to recover from acute injury, which involves specific phenotypic changes, but also significant profibrotic tubule-interstitial crosstalk. Tubule-derived profibrotic stimuli and subsequent myofibroblast activation and extracellular matrix deposition have been linked closely with decline of renal function and nephron loss. However, recent data have questioned the view of purely detrimental effects of myofibroblast activation in the injured kidney and even suggested its beneficial role for epithelial regeneration. This article reviews the current understanding of the underlying mechanisms of tubular cell turnover, new suggested pathways of proregenerative tubular-interstitial crosstalk, and relevant insights of proliferation-enhancing effects of myofibroblasts on epithelial cells in nonrenal tissues.
Collapse
|
15
|
Hanudel MR, Wong S, Jung G, Qiao B, Gabayan V, Zuk A, Ganz T. Amelioration of chronic kidney disease-associated anemia by vadadustat in mice is not dependent on erythroferrone. Kidney Int 2021; 100:79-89. [PMID: 33811979 DOI: 10.1016/j.kint.2021.03.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 02/02/2021] [Accepted: 03/10/2021] [Indexed: 02/06/2023]
Abstract
Vadadustat is an investigational hypoxia-inducible factor prolyl hydroxylase inhibitor that increases endogenous erythropoietin production and has been shown to decrease hepcidin levels, ameliorate iron restriction, and increase hemoglobin concentrations in anemic patients with chronic kidney disease (CKD). In studies of physiological responses to other erythropoietic stimuli, erythropoietin induced erythroblast secretion of erythroferrone (ERFE), which acts on the liver to suppress hepcidin production and mobilize iron for erythropoiesis. We therefore investigated whether vadadustat effects on erythropoiesis and iron metabolism are dependent on ERFE. Wild type and ERFE knockout mice with and without CKD were treated with vadadustat or vehicle. In both wild type and ERFE knockout CKD models, vadadustat was similarly effective, as evidenced by normalized hemoglobin concentrations, increased expression of duodenal iron transporters, lower serum hepcidin levels, and decreased tissue iron concentrations. This is consistent with ERFE-independent increased iron mobilization. Vadadustat treatment also lowered serum urea nitrogen and creatinine concentrations and decreased expression of kidney fibrosis markers. Lastly, vadadustat affected fibroblast growth factor 23 (FGF23) profiles: in non-CKD mice, vadadustat increased plasma total FGF23 out of proportion to intact FGF23, consistent with the known effects of hypoxia-inducible factor-1α and erythropoietin on FGF23 production and metabolism. However, in the mice with CKD, vadadustat markedly decreased both total and intact FGF23, effects likely contributed to by the reduced loss of kidney function. Thus, in this CKD model, vadadustat ameliorated anemia independently of ERFE, improved kidney parameters, and decreased FGF23. How vadadustat affects CKD progression in humans warrants future studies.
Collapse
Affiliation(s)
- Mark R Hanudel
- Department of Pediatrics, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA.
| | - Shirley Wong
- Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA
| | - Grace Jung
- Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA
| | - Bo Qiao
- Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA
| | - Victoria Gabayan
- Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA
| | - Anna Zuk
- Research and Development, Akebia Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Tomas Ganz
- Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
16
|
Qin LY, Lin X, Liu J, Dong R, Yuan J, Zha Y. The combination of vitamin D3 and erythropoietin alleviates acute kidney injury induced by ischemia-reperfusion via inhibiting inflammation and apoptosis. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:167-174. [PMID: 33953855 PMCID: PMC8061330 DOI: 10.22038/ijbms.2020.51384.11661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 12/12/2020] [Indexed: 01/20/2023]
Abstract
OBJECTIVES Acute renal ischemia may cause acute renal dysfunction due to lack of blood supply; the manifestations are renal tubular cell apoptosis, infiltration of macrophages, and microvascular destruction. Many studies have shown that erythropoietin (EPO) and vitamin D3 (VD3) can be used to prevent or treat renal ischemia-reperfusion (I/R) injury, and VD3 may interact with EPO. In the present study, the effects of the combination of VD3 and EPO in I/R acute kidney injury were studied. MATERIALS AND METHODS Rats were divided into 5 groups: sham-operated (SHAM), AKI without treatment (AKI-control), AKI treatment with VD3(AKI+VD3), AKI treatment with EPO(AKI+EPO), AKI treatment with VD3 and EPO(AKI+VD3+EPO). The effects of the combination of VD3 and EPO on AKI were assessed by histologic, inflammation, and apoptosis studies. RESULTS The degree of damage in renal tissue was significantly reduced in VD3, EPO, and combined groups. Combination therapy with VD3 and EPO markedly improved Creatinine clearance rate (CCr). The combined treatment group showed the lowest F4/80+ and CD68+ expressions. The expression of Bcl-2 in the combined treatment group was higher than those in VD3 group and the EPO group, while Bax's expression goes in the opposite direction. CONCLUSION This provides further evidence that VD3 and EPO have beneficial effects in I/R injury via anti-inflammatory and anti-apoptosis pathways. The synergistic protective effect of VD3 and EPO is of profound significance in the development of new strategies for the prevention and treatment of acute kidney injury (AKI).
Collapse
Affiliation(s)
- Long-yan Qin
- Department of Nephrology, Guizhou Provincial People’s Hospital & NHC Key Laboratory of Pulmonary Immunological Disease (Guizhou Provincial People’s Hospital), Guiyang, Guizhou, 550002, P.R. China
| | - Xin Lin
- Department of Nephrology, Guizhou Provincial People’s Hospital & NHC Key Laboratory of Pulmonary Immunological Disease (Guizhou Provincial People’s Hospital), Guiyang, Guizhou, 550002, P.R. China
| | - Juan Liu
- Department of Operating Room, The First Affiliated Hospital of Guizhou University of traditional Chinese medicine, Guiyang, Guizhou, 550001, P.R. China
| | - Rong Dong
- Department of Nephrology, Guizhou Provincial People’s Hospital & NHC Key Laboratory of Pulmonary Immunological Disease (Guizhou Provincial People’s Hospital), Guiyang, Guizhou, 550002, P.R. China
| | - Jing Yuan
- Department of Nephrology, Guizhou Provincial People’s Hospital & NHC Key Laboratory of Pulmonary Immunological Disease (Guizhou Provincial People’s Hospital), Guiyang, Guizhou, 550002, P.R. China
| | - Yan Zha
- Department of Nephrology, Guizhou Provincial People’s Hospital & NHC Key Laboratory of Pulmonary Immunological Disease (Guizhou Provincial People’s Hospital), Guiyang, Guizhou, 550002, P.R. China
| |
Collapse
|
17
|
Oshima M, Hara A, Toyama T, Jun M, Pollock C, Jardine M, Harrap S, Poulter N, Cooper ME, Woodward M, Chalmers J, Perkovic V, Wong MG, Wada T. Comparison of Circulating Biomarkers in Predicting Diabetic Kidney Disease Progression With Autoantibodies to Erythropoietin Receptor. Kidney Int Rep 2020; 6:284-295. [PMID: 33615053 PMCID: PMC7879109 DOI: 10.1016/j.ekir.2020.10.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/03/2020] [Accepted: 10/27/2020] [Indexed: 01/21/2023] Open
Abstract
Introduction Several circulating markers, including autoantibodies to erythropoietin receptor (anti-EPOR antibodies), have been identified as useful biomarkers in predicting diabetic kidney disease progression. However, a direct comparison of their utility is lacking. We aimed to validate and to compare the prognostic value of anti-EPOR antibodies with that of other known biomarkers, using the ADVANCE trial and its long-term follow-up, ADVANCE-ON, cohorts. Methods In this nested case-control study from the ADVANCE trial cohort, we included 165 case participants who had the composite kidney outcome (renal replacement therapy, renal death, or doubling of serum creatinine to ≥200 μmol/l) and 330 matched controls. We compared the associations of baseline plasma levels of anti-EPOR antibodies, tumor necrosis factor receptor (TNFR)-1 and -2, and bone morphogenetic protein (BMP)-7 with kidney outcomes. Results Cases had higher baseline plasma levels of anti-EPOR antibodies than controls (median 1.7 vs. 0.6 enzyme-linked immunosorbent assay unit, P < 0.001). Higher levels of anti-EPOR antibodies were associated with an increased risk of kidney outcome (odds ratio 2.16 [95% confidence interval 1.51, 3.08], per 1 SD of log-transformed levels) after adjusting for conventional markers. Elevated circulating TNFR1 and TNFR2 levels, and lower BMP-7 levels at baseline, were associated with poor kidney outcome (odds ratios 2.06 [1.29, 3.30], 1.66 [1.13, 2.43], and 0.45 [0.32, 0.65], respectively). The addition of anti-EPOR antibodies into the model improved the prediction of kidney outcome, regardless of other biomarkers. Conclusion Anti-EPOR antibodies provide a promising biomarker, as with TNFR1, TNFR2, and BMP-7, in predicting kidney disease progression in people with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Megumi Oshima
- Department of Renal and Metabolic, The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia.,Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan.,Renal Department, Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Akinori Hara
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Tadashi Toyama
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Min Jun
- Department of Renal and Metabolic, The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Carol Pollock
- Renal Department, Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Meg Jardine
- Department of Renal and Metabolic, The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia.,Nephrology Unit, Concord Repatriation General Hospital, Sydney, New South Wales, Australia
| | - Stephen Harrap
- Department of Physiology, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Neil Poulter
- International Center for Circulatory Health, Imperial College, London, UK
| | - Mark E Cooper
- Departiment of Diabetes, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Mark Woodward
- Department of Renal and Metabolic, The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia.,The George Institute for Global Health, University of Oxford, Oxford, UK.,Department of Epidemiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - John Chalmers
- Department of Renal and Metabolic, The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Vlado Perkovic
- Department of Renal and Metabolic, The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Muh Geot Wong
- Department of Renal and Metabolic, The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia.,Renal Department, Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Takashi Wada
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
18
|
Peritubular Capillary Rarefaction: An Underappreciated Regulator of CKD Progression. Int J Mol Sci 2020; 21:ijms21218255. [PMID: 33158122 PMCID: PMC7662781 DOI: 10.3390/ijms21218255] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 10/29/2020] [Indexed: 12/15/2022] Open
Abstract
Peritubular capillary (PTC) rarefaction is commonly detected in chronic kidney disease (CKD) such as hypertensive nephrosclerosis and diabetic nephropathy. Moreover, PTC rarefaction prominently correlates with impaired kidney function and predicts the future development of end-stage renal disease in patients with CKD. However, it is still underappreciated that PTC rarefaction is a pivotal regulator of CKD progression, primarily because the molecular mechanisms of PTC rarefaction have not been well-elucidated. In addition to the established mechanisms (reduced proangiogenic factors and increased anti-angiogenic factors), recent studies discovered significant contribution of the following elements to PTC loss: (1) prompt susceptibility of PTC to injury, (2) impaired proliferation of PTC, (3) apoptosis/senescence of PTC, and (4) pericyte detachment from PTC. Mainly based on the recent and novel findings in basic research and clinical study, this review describes the roles of the above-mentioned elements in PTC loss and focuses on the major factors regulating PTC angiogenesis, the assessment of PTC rarefaction and its surrogate markers, and an overview of the possible therapeutic agents to mitigate PTC rarefaction during CKD progression. PTC rarefaction is not only a prominent histological characteristic of CKD but also a central driving force of CKD progression.
Collapse
|
19
|
Liu B, Tan P. PPAR γ/TLR4/TGF-β1 axis mediates the protection effect of erythropoietin on cyclosporin A-induced chronic nephropathy in rat. Ren Fail 2020; 42:216-224. [PMID: 32090669 PMCID: PMC7054967 DOI: 10.1080/0886022x.2020.1729188] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/18/2019] [Accepted: 02/04/2020] [Indexed: 01/12/2023] Open
Abstract
Objective: Nephrotoxicity is the main side effect of cyclosporine A and finding an effective combating method is urgent. The present study investigates the improving effect of erythropoietin (EPO) on cyclosporine A induce renal injury in rats and further explores its possible mechanism.Methods: Recombinant adenovirus for expression of EPO was constructed and injected into kidney with multipoint. Levels of blood urea nitrogen (BUN) and serum creatinine (SCr) were detected by kits. HE staining and Masson's trichrome staining were used to evaluate pathological changes. ELISA was performed to detect the levels of transforming growth factor (TGF)-β1, interleukin (IL)-1β, and IL-6 in serum. Levels of malondialdehyde (MDA) and superoxide dismutase (SOD) in kidney were detected according to manufacturer's instruction. Western blotting was performed to observe the protein expression levels of peroxisome proliferator-activated receptor γ (PPAR γ), Toll-like receptor (TLR) 4, and TGF-β1.Results: Results showed that EPO overexpression in rat kidney could significantly improve renal injury and fibrosis, suppress the release of inflammatory factors and reduce oxidative stress induced by cyclosporine A. Western blotting results showed that EPO overexpression could up-regulate the expression of PPARγ and down-regulate the expression of TLR4 and TGF-β1. Interestingly, when PPARγ activity was inhibited by T0070907, an effective and specific PPARγ inhibitor, the therapeutic effect of EPO was significantly attenuated.Conclusion: Taken together, above results shown the protective effect of EPO on cyclosporine A-induced renal injury and confirmed that EPO's anti-inflammation and antioxidative stress involving the PPAR γ/TLR4/TGFβ1 axis.
Collapse
Affiliation(s)
- Bin Liu
- Department of Nephrology and Rheumatology, Chinese Medicine Hospital of Hainan Province, Haikou, China
| | - Ping Tan
- Department of Nephrology and Rheumatology, Chinese Medicine Hospital of Hainan Province, Haikou, China
| |
Collapse
|
20
|
Maique J, Flores B, Shi M, Shepard S, Zhou Z, Yan S, Moe OW, Hu MC. High Phosphate Induces and Klotho Attenuates Kidney Epithelial Senescence and Fibrosis. Front Pharmacol 2020; 11:1273. [PMID: 32973510 PMCID: PMC7468469 DOI: 10.3389/fphar.2020.01273] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 07/31/2020] [Indexed: 12/18/2022] Open
Abstract
Cellular senescence is an irreversible cell growth arrest and is associated with aging and age-related diseases. High plasma phosphate (Pi) and deficiency of Klotho contribute to aging and kidney fibrosis, a pathological feature in the aging kidney and chronic kidney disease. This study examined the interactive role of Pi and Klotho in kidney senescence and fibrosis. Homozygous Klotho hypomorphic mice had high plasma Pi, undetectable Klotho in plasma and kidney, high senescence with massive collagen accumulation in kidney tubules, and fibrin deposits in peritubular capillaries. To examine the Pi effect on kidney senescence, a high (2%) Pi diet was given to wild-type mice. One week of high dietary Pi mildly increased plasma Pi, and upregulated kidney p16/p21 expression, but did not significantly decrease Klotho. Two weeks of high Pi intake led to increase in plasminogen activator inhibitor (PAI)-1, and decrease in kidney Klotho, but still without detectable increase in kidney fibrosis. More prolonged dietary Pi for 12 weeks exacerbated kidney senescence and fibrosis; more so in heterozygous Klotho hypomorphic mice compared to wild-type mice, and in mice with chronic kidney disease (CKD) on high Pi diet compared to CKD mice fed a normal Pi diet. In cultured kidney tubular cells, high Pi directly induced cellular senescence, injury and epithelial-mesenchymal transition, and enhanced H2O2-induced cellular senescence and injury, which were abrogated by Klotho. Fucoidan, a bioactive molecule with multiple biologic functions including senescence inhibition, blunted Pi-induced cellular senescence, oxidation, injury, epithelial-mesenchymal transition, and senescence-associated secretary phenotype. In conclusion, high Pi activates senescence through distinct but interconnected mechanisms: upregulating p16/p21 (early), and elevating plasminogen activator inhibitor-1 and downregulating Klotho (late). Klotho may be a promising agent to attenuate senescence and ameliorate age-associated, and Pi-induced kidney degeneration such as kidney fibrosis.
Collapse
Affiliation(s)
- Jenny Maique
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Brianna Flores
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Mingjun Shi
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Sierra Shepard
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Zhiyong Zhou
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Shirely Yan
- Departments of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
21
|
Wu Y, Chen W, Zhang Y, Liu A, Yang C, Wang H, Zhu T, Fan Y, Yang B. Potent Therapy and Transcriptional Profile of Combined Erythropoietin-Derived Peptide Cyclic Helix B Surface Peptide and Caspase-3 siRNA against Kidney Ischemia/Reperfusion Injury in Mice. J Pharmacol Exp Ther 2020; 375:92-103. [PMID: 32759272 DOI: 10.1124/jpet.120.000092] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/27/2020] [Indexed: 12/19/2022] Open
Abstract
Cause-specific treatment and timely diagnosis are still not available for acute kidney injury (AKI) apart from supportive therapy and serum creatinine measurement. A novel erythropoietin-derived cyclic helix B surface peptide (CHBP) protects kidneys against AKI with different causes, but the underlying mechanism is not fully defined. Herein, we investigated the transcriptional profile of renoprotection induced by CHBP and its potential synergistic effects with siRNA targeting caspase-3, an executing enzyme of apoptosis and inflammation (CASP3siRNA), on ischemia/reperfusion (IR)-induced AKI. Utilizing a mouse model with 30-minute renal bilateral ischemia and 48-hour reperfusion, the renoprotection of CHBP or CASP3siRNA was demonstrated in renal function and structure, active caspase-3 and HMGB1 expression. Combined treatment of CHBP and CASP3siRNA further preserved kidney structure and reduced active caspase-3 and HMGB1. Furthermore, differentially expressed genes (DEGs) were identified with fold change >1.414 and P < 0.05. In IR kidneys, 281 DEGs induced by CHBP were mainly involved in promoting cell division and improving cellular function and metabolism (upregulated signal transducer and activator of transcription 5B and solute carrier family 22 member 7). The additional administration of CASP3siRNA caused 504 and 418 DEGs in IR + CHBP kidneys with or without negative control small-interfering RNA, with 37 genes in common. These DEGs were associated with modulated apoptosis and inflammation (upregulated BCL6, SLPI, and SERPINA3M) as well as immunity, injury, and microvascular homeostasis (upregulated complement factor H and GREM1 and downregulated ANGPTL2). This proof-of-effect study indicated the potent renoprotection of CASP3siRNA upon CHBP at the early stage of IR-induced AKI. Underlying genes, BCL6, SLPI, SERPINA3M, GREM1, and ANGPTL2, might be potential new biomarkers for clinical applications. SIGNIFICANCE STATEMENT: It is imperative to explore new strategies of cause-specific treatment and timely diagnosis for acute kidney injury (AKI). CHBP and CASP3siRNA synergistically protected kidney structure after 48-hour ischemia/reperfusion-induced AKI with reduced injury mediators CASP3 and high mobility group box 1. CHBP upregulated cell division-, function-, and metabolism-related genes, whereas CASP3siRNA further regulated immune response- and tissue homeostasis-associated genes. Combined CHBP and CASP3siRNA might be a potent and specific treatment for AKI, and certain dysregulated genes secretory leukocyte peptidase inhibitor and SERPINA3M could facilitate timely diagnosis.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Renal Group, Basic Medical Research Centre, Nantong University, Nantong, China (Y.W., Y.Z., A.L.); Leicester-Nantong Joint Institute of Kidney Science, Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China (W.C., H.W., Y.F., B.Y.); Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China (C.Y., T.Z.); Shanghai Key Laboratory of Organ Transplantation, Shanghai, China (C.Y., T.Z.); and Renal Group, Department of Cardiovascular Sciences, University of Leicester, University Hospitals of Leicester, Leicester, United Kingdom (Y.W., B.Y.)
| | - Weiwei Chen
- Renal Group, Basic Medical Research Centre, Nantong University, Nantong, China (Y.W., Y.Z., A.L.); Leicester-Nantong Joint Institute of Kidney Science, Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China (W.C., H.W., Y.F., B.Y.); Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China (C.Y., T.Z.); Shanghai Key Laboratory of Organ Transplantation, Shanghai, China (C.Y., T.Z.); and Renal Group, Department of Cardiovascular Sciences, University of Leicester, University Hospitals of Leicester, Leicester, United Kingdom (Y.W., B.Y.)
| | - Yufang Zhang
- Renal Group, Basic Medical Research Centre, Nantong University, Nantong, China (Y.W., Y.Z., A.L.); Leicester-Nantong Joint Institute of Kidney Science, Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China (W.C., H.W., Y.F., B.Y.); Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China (C.Y., T.Z.); Shanghai Key Laboratory of Organ Transplantation, Shanghai, China (C.Y., T.Z.); and Renal Group, Department of Cardiovascular Sciences, University of Leicester, University Hospitals of Leicester, Leicester, United Kingdom (Y.W., B.Y.)
| | - Aifen Liu
- Renal Group, Basic Medical Research Centre, Nantong University, Nantong, China (Y.W., Y.Z., A.L.); Leicester-Nantong Joint Institute of Kidney Science, Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China (W.C., H.W., Y.F., B.Y.); Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China (C.Y., T.Z.); Shanghai Key Laboratory of Organ Transplantation, Shanghai, China (C.Y., T.Z.); and Renal Group, Department of Cardiovascular Sciences, University of Leicester, University Hospitals of Leicester, Leicester, United Kingdom (Y.W., B.Y.)
| | - Cheng Yang
- Renal Group, Basic Medical Research Centre, Nantong University, Nantong, China (Y.W., Y.Z., A.L.); Leicester-Nantong Joint Institute of Kidney Science, Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China (W.C., H.W., Y.F., B.Y.); Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China (C.Y., T.Z.); Shanghai Key Laboratory of Organ Transplantation, Shanghai, China (C.Y., T.Z.); and Renal Group, Department of Cardiovascular Sciences, University of Leicester, University Hospitals of Leicester, Leicester, United Kingdom (Y.W., B.Y.)
| | - Hui Wang
- Renal Group, Basic Medical Research Centre, Nantong University, Nantong, China (Y.W., Y.Z., A.L.); Leicester-Nantong Joint Institute of Kidney Science, Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China (W.C., H.W., Y.F., B.Y.); Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China (C.Y., T.Z.); Shanghai Key Laboratory of Organ Transplantation, Shanghai, China (C.Y., T.Z.); and Renal Group, Department of Cardiovascular Sciences, University of Leicester, University Hospitals of Leicester, Leicester, United Kingdom (Y.W., B.Y.)
| | - Tongyu Zhu
- Renal Group, Basic Medical Research Centre, Nantong University, Nantong, China (Y.W., Y.Z., A.L.); Leicester-Nantong Joint Institute of Kidney Science, Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China (W.C., H.W., Y.F., B.Y.); Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China (C.Y., T.Z.); Shanghai Key Laboratory of Organ Transplantation, Shanghai, China (C.Y., T.Z.); and Renal Group, Department of Cardiovascular Sciences, University of Leicester, University Hospitals of Leicester, Leicester, United Kingdom (Y.W., B.Y.)
| | - Yaping Fan
- Renal Group, Basic Medical Research Centre, Nantong University, Nantong, China (Y.W., Y.Z., A.L.); Leicester-Nantong Joint Institute of Kidney Science, Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China (W.C., H.W., Y.F., B.Y.); Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China (C.Y., T.Z.); Shanghai Key Laboratory of Organ Transplantation, Shanghai, China (C.Y., T.Z.); and Renal Group, Department of Cardiovascular Sciences, University of Leicester, University Hospitals of Leicester, Leicester, United Kingdom (Y.W., B.Y.)
| | - Bin Yang
- Renal Group, Basic Medical Research Centre, Nantong University, Nantong, China (Y.W., Y.Z., A.L.); Leicester-Nantong Joint Institute of Kidney Science, Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China (W.C., H.W., Y.F., B.Y.); Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China (C.Y., T.Z.); Shanghai Key Laboratory of Organ Transplantation, Shanghai, China (C.Y., T.Z.); and Renal Group, Department of Cardiovascular Sciences, University of Leicester, University Hospitals of Leicester, Leicester, United Kingdom (Y.W., B.Y.)
| |
Collapse
|
22
|
Abstract
Metabolic disorders, such as diabetes mellitus (DM), are increasingly becoming significant risk factors for the health of the global population and consume substantial portions of the gross domestic product of all nations. Although conventional therapies that include early diagnosis, nutritional modification of diet, and pharmacological treatments may limit disease progression, tight serum glucose control cannot prevent the onset of future disease complications. With these concerns, novel strategies for the treatment of metabolic disorders that involve the vitamin nicotinamide, the mechanistic target of rapamycin (mTOR), mTOR Complex 1 (mTORC1), mTOR Complex 2 (mTORC2), AMP activated protein kinase (AMPK), and the cellular pathways of autophagy and apoptosis offer exceptional promise to provide new avenues of treatment. Oversight of these pathways can promote cellular energy homeostasis, maintain mitochondrial function, improve glucose utilization, and preserve pancreatic beta-cell function. Yet, the interplay among mTOR, AMPK, and autophagy pathways can be complex and affect desired clinical outcomes, necessitating further investigations to provide efficacious treatment strategies for metabolic dysfunction and DM.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, New York 10022,
| |
Collapse
|
23
|
Li P, Shi M, Maique J, Shaffer J, Yan S, Moe OW, Hu MC. Beclin 1/Bcl-2 complex-dependent autophagy activity modulates renal susceptibility to ischemia-reperfusion injury and mediates renoprotection by Klotho. Am J Physiol Renal Physiol 2020; 318:F772-F792. [PMID: 31984794 PMCID: PMC7099499 DOI: 10.1152/ajprenal.00504.2019] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/10/2020] [Accepted: 01/16/2020] [Indexed: 02/06/2023] Open
Abstract
Klotho- and beclin 1-driven autophagy extends life. We examined the role of beclin 1 in modifying acute kidney injury (AKI) and whether beclin 1 mediates Klotho's known renoprotective action in AKI. AKI was induced by ischemia-reperfusion injury in mice with different levels of autophagy activity by genetic manipulation: wild-type (WT) mice with normal beclin 1 expression and function, mice with normal beclin 1 levels but high activity through knockin of gain-of-function mutant beclin 1 (Becn1F121A), mice with low beclin 1 levels and activity caused by heterozygous global deletion of beclin 1 (Becn1+/-), or mice with extremely low beclin 1 activity from knockin of the mutant constitutively active beclin 1 inhibitor Bcl-2 (Bcl2AAA). Klotho was increased by transgenic overexpression (Tg-Kl) or recombinant Klotho protein administration. After ischemia-reperfusion injury, Becn1F121A mice (high autophagy) had milder AKI and Becn1+/- and Bcl2AAA mice (low autophagy) had more severe AKI than WT mice. Tg-Kl mice had milder AKI, but its renoprotection was partially attenuated in Becn1+/-;Tg-Kl mice and was significantly reduced, although not completely abolished, in Bcl2AAA;Tg-Kl mice. Recombinant Klotho protein conferred more renoprotection from AKI in WT mice than in Becn1+/- or Bcl2AAA mice. Klotho reduced beclin 1/Bcl-2 protein complexes and increased autophagy activity, but this effect was less prominent in mice or cells with Bcl2AAA. Transfected Bcl2AAA or Becn1F123A decreased or increased autophagy activity and rendered cells more susceptible or more resistant to oxidative cytotoxicity, respectively. In conclusion, beclin 1 confers renoprotection by activating autophagy. Klotho protects the kidney partially via disruption of beclin 1/Bcl-2 interactions and enhancement of autophagy activity.
Collapse
Affiliation(s)
- Peng Li
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Mingjun Shi
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jenny Maique
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Joy Shaffer
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Shirley Yan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
24
|
Shi M, Maique J, Shaffer J, Davidson T, Sebti S, Fernández ÁF, Zou Z, Yan S, Levine B, Moe OW, Hu MC. The tripartite interaction of phosphate, autophagy, and αKlotho in health maintenance. FASEB J 2020; 34:3129-3150. [PMID: 31908069 PMCID: PMC7286356 DOI: 10.1096/fj.201902127r] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 12/19/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022]
Abstract
Aging-related organ degeneration is driven by multiple factors including the cell maintenance mechanisms of autophagy, the cytoprotective protein αKlotho, and the lesser known effects of excess phosphate (Pi), or phosphotoxicity. To examine the interplay between Pi, autophagy, and αKlotho, we used the BK/BK mouse (homozygous for mutant Becn1F121A ) with increased autophagic flux, and αKlotho-hypomorphic mouse (kl/kl) with impaired urinary Pi excretion, low autophagy, and premature organ dysfunction. BK/BK mice live longer than WT littermates, and have heightened phosphaturia from downregulation of two key NaPi cotransporters in the kidney. The multi-organ failure in kl/kl mice was rescued in the double-mutant BK/BK;kl/kl mice exhibiting lower plasma Pi, improved weight gain, restored plasma and renal αKlotho levels, decreased pathology of multiple organs, and improved fertility compared to kl/kl mice. The beneficial effects of heightened autophagy from Becn1F121A was abolished by chronic high-Pi diet which also shortened life span in the BK/BK;kl/kl mice. Pi promoted beclin 1 binding to its negative regulator BCL2, which impairs autophagy flux. Pi downregulated αKlotho, which also independently impaired autophagy. In conclusion, Pi, αKlotho, and autophagy interact intricately to affect each other. Both autophagy and αKlotho antagonizes phosphotoxicity. In concert, this tripartite system jointly determines longevity and life span.
Collapse
Affiliation(s)
- Mingjun Shi
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jenny Maique
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joy Shaffer
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Taylor Davidson
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Salwa Sebti
- Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Álvaro F. Fernández
- Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zhongju Zou
- Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shirley Yan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Beth Levine
- Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Orson W. Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|