1
|
Khan MZ, Zugaza JL, Torres Aleman I. The signaling landscape of insulin-like growth factor 1. J Biol Chem 2025; 301:108047. [PMID: 39638246 PMCID: PMC11748690 DOI: 10.1016/j.jbc.2024.108047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
The sheer amplitude of biological actions of insulin-like growth factor I (IGF-1) affecting all types of cells in all tissues suggests a vast signaling landscape for this ubiquitous humoral signal. While the canonical signaling pathways primarily involve the Ras/MAPK and PI3K/AKT cascades, the evolutionary conservation of insulin-like peptides (ILPs) and their pathways hints at the potential for novel functions to emerge over time. Indeed, the evolutionary trajectory of ILPs opens the possibility of either novel functions for these two pathways, novel downstream routes, or both. Evidence supporting this notion includes observations of neofunctionalization in bony fishes or crustaceans, and the involvement of ILPs pathways in invertebrate eusociality or in vertebrate bone physiology, respectively. Such evolutionary processes likely contribute to the rich diversity of ILPs signaling observed today. Moreover, the interplay between conserved signaling pathways, such as those implicated in aging (predominantly involving the PI3K-AKT route), and lesser known pathways, such as those mediated by biased G-protein coupled receptors and others even less known, may underpin the context-dependent actions characteristic of ILPs signaling. While canonical IGF-1 signaling is often assumed to account for the intracellular pathways utilized by this growth factor, a comprehensive analysis of all the pathways mediated by the IGF-1 receptor (IGF-1R) remains lacking. This review aims to explore both canonical and non-canonical routes of IGF-1R action across various cell types, offering a detailed examination of the mechanisms underlying IGF-1 signaling and highlighting the significant gaps in our current understanding.
Collapse
Affiliation(s)
- Muhammad Zahid Khan
- Achucarro Basque Center for Neuroscience, Leioa, Spain; CIBERNED, Madrid, Spain
| | - Jose Luis Zugaza
- Achucarro Basque Center for Neuroscience, Leioa, Spain; Ikerbasque Science Foundation, Bilbao, Spain
| | - Ignacio Torres Aleman
- Achucarro Basque Center for Neuroscience, Leioa, Spain; CIBERNED, Madrid, Spain; Ikerbasque Science Foundation, Bilbao, Spain.
| |
Collapse
|
2
|
Lay AC, Tran VDT, Nair V, Betin V, Hurcombe JA, Barrington AF, Pope RJ, Burdet F, Mehl F, Kryvokhyzha D, Ahmad A, Sinton MC, Lewis P, Wilson MC, Menon R, Otto E, Heesom KJ, Ibberson M, Looker HC, Nelson RG, Ju W, Kretzler M, Satchell SC, Gomez MF, Coward RJM. Profiling of insulin-resistant kidney models and human biopsies reveals common and cell-type-specific mechanisms underpinning Diabetic Kidney Disease. Nat Commun 2024; 15:10018. [PMID: 39562547 PMCID: PMC11576882 DOI: 10.1038/s41467-024-54089-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 11/01/2024] [Indexed: 11/21/2024] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of end stage kidney failure worldwide, of which cellular insulin resistance is a major driver. Here, we study key human kidney cell types implicated in DKD (podocytes, glomerular endothelial, mesangial and proximal tubular cells) in insulin sensitive and resistant conditions, and perform simultaneous transcriptomics and proteomics for integrated analysis. Our data is further compared with bulk- and single-cell transcriptomic kidney biopsy data from early- and advanced-stage DKD patient cohorts. We identify several consistent changes (individual genes, proteins, and molecular pathways) occurring across all insulin-resistant kidney cell types, together with cell-line-specific changes occurring in response to insulin resistance, which are replicated in DKD biopsies. This study provides a rich data resource to direct future studies in elucidating underlying kidney signalling pathways and potential therapeutic targets in DKD.
Collapse
Affiliation(s)
- Abigail C Lay
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Van Du T Tran
- Vital-IT group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Viji Nair
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Virginie Betin
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | | | | | - Robert Jp Pope
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Frédéric Burdet
- Vital-IT group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Florence Mehl
- Vital-IT group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Dmytro Kryvokhyzha
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Abrar Ahmad
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Matthew C Sinton
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Philip Lewis
- Proteomics Facility, University of Bristol, Bristol, UK
| | | | - Rajasree Menon
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Edgar Otto
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Kate J Heesom
- Proteomics Facility, University of Bristol, Bristol, UK
| | - Mark Ibberson
- Vital-IT group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Helen C Looker
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institute of Health, Phoenix, AZ, USA
| | - Robert G Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institute of Health, Phoenix, AZ, USA
| | - Wenjun Ju
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Simon C Satchell
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Maria F Gomez
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Richard J M Coward
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK.
| |
Collapse
|
3
|
Chen F, Cai X, Yu Y. PHB2 alleviates retinal pigment epithelium cell fibrosis by suppressing the AGE-RAGE pathway. Open Life Sci 2024; 19:20220985. [PMID: 39507806 PMCID: PMC11538926 DOI: 10.1515/biol-2022-0985] [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: 05/28/2024] [Revised: 09/03/2024] [Accepted: 09/23/2024] [Indexed: 11/08/2024] Open
Abstract
Fibrosis is the primary cause of retinal detachment and visual decline. Here, we investigated the role of Prohibitin 2 (PHB2) in modulating fibrosis in ARPE-19 cells stimulated by transforming growth factor (TGF)-β2. The proliferation, migration, and apoptosis of ARPE-19 cells were evaluated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, wound healing, and flow cytometry assays, and levels of fibrosis-associated and pathway-related proteins were determined by performing western blotting. To examine the mechanisms underlying ARPE-19 cell fibrosis, we performed RNA sequencing, protein-protein interaction network, and enrichment analyses. We detected increases in the expression of the fibrosis-related proteins fibronectin and collagen I in response to TGF-β2 treatment, whereas the expression of PHB2 was downregulated. PHB2 overexpression suppressed the proliferation and migration of TGF-β2-stimulated ARPE-19 cells, promoted apoptosis, and inhibited fibrosis and Smad and non-Smad pathways. PHB2 overexpression inhibited the advanced glycation end-product (AGE)-receptor of advanced glycation end-product (RAGE) pathway activated by TGF-β2 treatment, which contributed to enhancing the effects of PHB2 on cellular processes, fibrosis, and Smad and non-Smad pathways. Conversely, exogenous application of AGE counteracted the effects of PHB2 overexpression. We conclude that by suppressing the AGE-RAGE pathway, PHB2 exerts an inhibitory effect on TGF-β2-induced fibrosis in ARPE-19 cells.
Collapse
Affiliation(s)
- Feng Chen
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, No. 9, Jinsui Road, Tianhe District, Guangzhou, Guangdong, 510623, China
| | - Xiaoxiao Cai
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, China
| | - Ying Yu
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, China
| |
Collapse
|
4
|
Hurcombe JA, Barrington F, Marchetti M, Betin VM, Bowen EE, Lay AC, Ni L, Dayalan L, Pope RJ, Brinkkoetter PT, Holzenberger M, Welsh GI, Coward RJ. Contrasting consequences of podocyte insulin-like growth factor 1 receptor inhibition. iScience 2024; 27:109749. [PMID: 38706850 PMCID: PMC11068853 DOI: 10.1016/j.isci.2024.109749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/12/2023] [Accepted: 04/11/2024] [Indexed: 05/07/2024] Open
Abstract
Insulin signaling to the glomerular podocyte via the insulin receptor (IR) is critical for kidney function. In this study we show that near-complete knockout of the closely related insulin-like growth factor 1 receptor (IGF1R) in podocytes is detrimental, resulting in albuminuria in vivo and podocyte cell death in vitro. In contrast, partial podocyte IGF1R knockdown confers protection against doxorubicin-induced podocyte injury. Proteomic analysis of cultured podocytes revealed that while near-complete loss of podocyte IGF1R results in the downregulation of mitochondrial respiratory complex I and DNA damage repair proteins, partial IGF1R inhibition promotes respiratory complex expression. This suggests that altered mitochondrial function and resistance to podocyte stress depends on the level of IGF1R suppression, the latter determining whether receptor inhibition is protective or detrimental. Our work suggests that the partial suppression of podocyte IGF1R could have therapeutic benefits in treating albuminuric kidney disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lan Ni
- Bristol Renal, University of Bristol, Bristol, UK
| | | | | | - Paul T. Brinkkoetter
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | | | | |
Collapse
|
5
|
Hejazian SM, Ardalan M, Hosseiniyan Khatibi SM, Rahbar Saadat Y, Barzegari A, Gueguen V, Meddahi-Pellé A, Anagnostou F, Zununi Vahed S, Pavon-Djavid G. Biofactors regulating mitochondrial function and dynamics in podocytes and podocytopathies. J Cell Physiol 2023; 238:2206-2227. [PMID: 37659096 DOI: 10.1002/jcp.31110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/25/2023] [Accepted: 08/14/2023] [Indexed: 09/04/2023]
Abstract
Podocytes are terminally differentiated kidney cells acting as the main gatekeepers of the glomerular filtration barrier; hence, inhibiting proteinuria. Podocytopathies are classified as kidney diseases caused by podocyte damage. Different genetic and environmental risk factors can cause podocyte damage and death. Recent evidence shows that mitochondrial dysfunction also contributes to podocyte damage. Understanding alterations in mitochondrial metabolism and function in podocytopathies and whether altered mitochondrial homeostasis/dynamics is a cause or effect of podocyte damage are issues that need in-depth studies. This review highlights the roles of mitochondria and their bioenergetics in podocytes. Then, factors/signalings that regulate mitochondria in podocytes are discussed. After that, the role of mitochondrial dysfunction is reviewed in podocyte injury and the development of different podocytopathies. Finally, the mitochondrial therapeutic targets are considered.
Collapse
Affiliation(s)
| | | | | | | | - Abolfazl Barzegari
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Villetaneuse, France
| | - Virginie Gueguen
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Villetaneuse, France
| | - Anne Meddahi-Pellé
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Villetaneuse, France
| | - Fani Anagnostou
- Université de Paris, CNRS UMR 7052 INSERM U1271, B3OA, Paris, France
| | | | - Graciela Pavon-Djavid
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Villetaneuse, France
| |
Collapse
|
6
|
Mani I, Singh V. An overview of receptor endocytosis and signaling. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 194:1-18. [PMID: 36631188 DOI: 10.1016/bs.pmbts.2022.06.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Endocytosis is a cellular process which mediates receptor internalization, nutrient uptake, and the regulation of cell signaling. Microorganisms (many bacteria and viruses) and toxins also use the same process and enter the cells. Generally, endocytosis is considered in the three forms such as phagocytosis (cell eating), pinocytosis (cell drinking), and highly selective receptor-mediated endocytosis (clathrin-dependent and independent). Several endocytic routes exist in an analogous, achieving diverse functions. Most studies on endocytosis have used transformed cells in culture. To visualize the receptor internalization, trafficking, and signaling in subcellular organelles, a green fluorescent protein-tagged receptor has been utilized. It also helps to visualize the endocytosis effects in live-cell imaging. Confocal laser microscopy increases our understanding of receptor endocytosis and signaling. Site-directed mutagenesis studies demonstrated that many short-sequence motifs of the cytoplasmic domain of receptors significantly play a vital role in receptor internalization, subcellular trafficking, and signaling. However, other factors also regulate receptor internalization through clathrin-coated vesicles. Receptor endocytosis can occur through clathrin-dependent and clathrin-independent pathways. This chapter briefly discusses the internalization, trafficking, and signaling of various receptors in normal conditions. In addition, it also highlights the malfunction of the receptor in disease conditions.
Collapse
Affiliation(s)
- Indra Mani
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India.
| | - Vijai Singh
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, Gujarat, India
| |
Collapse
|
7
|
Mani I, Singh V. Receptor biology: Challenges and opportunities. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:337-349. [PMID: 36813364 DOI: 10.1016/bs.pmbts.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Receptor biology provides a great opportunity to understand the ligand-receptor signaling involved in health and disease processes. Receptor endocytosis and signaling play a vital role in health conditions. Receptor-based signaling is the main form of communication between cells and cells with the environment. However, if any irregularities happen during these events, the consequences of pathophysiological conditions occur. Various methods are utilized to know structure, function, and regulation of receptor proteins. Further, live-cell imaging and genetic manipulations have aided in the understanding of receptor internalization, subcellular trafficking, signaling, metabolic degradation, etc. Understanding the genetics, biochemistry, and physiology of receptors and ligands is very helpful to explore various aspects such as prognosis, diagnosis, and treatment of disease. However, there are enormous challenges that exist to explore receptor biology further. This chapter briefly discusses the current challenges and emerging opportunities of receptor biology.
Collapse
Affiliation(s)
- Indra Mani
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India.
| | - Vijai Singh
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, Gujarat, India
| |
Collapse
|
8
|
Nonreceptor Tyrosine Kinase c-Abl-Mediated PHB2 Phosphorylation Aggravates Mitophagy Disorder in Parkinson’s Disease Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9233749. [PMID: 36406767 PMCID: PMC9668474 DOI: 10.1155/2022/9233749] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/13/2022] [Accepted: 10/13/2022] [Indexed: 11/10/2022]
Abstract
Mitophagy and oxidative stress play important roles in Parkinson's disease (PD). Dysregulated mitophagy exacerbates mitochondrial oxidative damage; however, the regulatory mechanism of mitophagy is unclear. Here, we provide a potential mechanistic link between c-Abl, a nonreceptor tyrosine kinase, and mitophagy in PD progression. We found that c-Abl activation reduces the interaction of prohibitin 2 (PHB2) and microtubule-associated protein 1 light chain 3 (LC3) and decreases the expressive level of antioxidative stress proteins, including nuclear factor erythroid 2-related factor 2 (Nrf2), NADPH quinone oxidoreductase-1 (NQO-1), and the antioxidant enzyme heme oxygenase-1 (HO-1) in 1-methyl-4-phenylpyridinium- (MPP+-) lesioned SH-SY5Y cells. Importantly, we found that MPP+ can increase the expression of phosphorylated proteins at the tyrosine site of PHB2 and the interaction of c-Abl with PHB2. We showed for the first time that PHB2 by changing tyrosine (Y) to aspartate (D) at site 121 resulted in impaired binding of PHB2 and LC3 in vitro. Moreover, silencing of PHB2 can decrease the interaction of PHB2 and LC3 and exacerbate the loss of dopaminergic neurons. We also found that STI 571, a c-Abl family kinase inhibitor, can decrease dopaminergic neuron damage and ameliorate MPTP-induced behavioral deficits in PD mice. Taken together, our findings highlight a novel molecular mechanism for aberrant PHB2 phosphorylation as an inhibitor of c-Abl activity and suggest that c-Abl and PHB2 are potential therapeutic targets for the treatment of individuals with PD. However, these results need to be further validated in PHB2 Y121D mice.
Collapse
|
9
|
Guo R, Wang P, Zheng X, Cui W, Shang J, Zhao Z. SGLT2 inhibitors suppress epithelial-mesenchymal transition in podocytes under diabetic conditions via downregulating the IGF1R/PI3K pathway. Front Pharmacol 2022; 13:897167. [PMID: 36225569 PMCID: PMC9550168 DOI: 10.3389/fphar.2022.897167] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 08/23/2022] [Indexed: 01/11/2023] Open
Abstract
Loss of podocyte is a characteristic pathological change of diabetic nephropathy (DN) which is associated with increased proteinuria. Many studies have shown that novel inhibitors of sodium-glucose cotransporter 2 (SGLT2-is), such as dapagliflozin, exert nephroprotective effect on delaying DN progression. However, the mechanisms underlying SGLT2-associated podocyte injury are still not fully elucidated. Here, we generated streptozotocin-induced DN models and treated them with dapagliflozin to explore the possible mechanisms underlying SGLT2 regulation. Compared to mice with DN, dapagliflozin-treated mice exhibited remission of pathological lesions, including glomerular sclerosis, thickening of the glomerular basement membrane (GBM), podocyte injury in the glomeruli, and decreased nephrotoxin levels accompanied by decreased SGLT2 expression. The mRNA expression profiles of these treated mice revealed the significance of the insulin-like growth factor-1 receptor (IGF1R)/PI3K regulatory axis in glomerular injury. KEGG analysis confirmed that the phosphatidylinositol signaling system and insulin signaling pathway were enriched. Western blotting showed that SGLT2-is inhibited the increase of mesenchymal markers (α-SMA, SNAI-1, and ZEB2) and the loss of podocyte markers (nephrin and E-cad). Additionally, SGLT2, IGF1R, phosphorylated PI3K, α-SMA, SNAI-1, and ZEB2 protein levels were increased in high glucose-stimulated human podocytes (HPC) and significantly decreased in dapagliflozin-treated (50 nM and 100 nM) or OSI-906-treated (inhibitor of IGF1R, 60 nM) groups. However, the use of both inhibitors did not enhance this protective effect. Next, we analyzed urine and plasma samples from a cohort consisting of 13 healthy people and 19 DN patients who were administered with (n = 9) or without (n = 10) SGLT2 inhibitors. ELISA results showed decreased circulating levels of IGF1 and IGF2 in SGLT2-is-treated DN patients compared with DN patients. Taken together, our study reported the key role of SGLT2/IGF1R/PI3K signaling in regulating podocyte epithelial-mesenchymal transition (EMT). Modulating IGF1R expression may be a novel approach for DN therapy.
Collapse
Affiliation(s)
- Ruixue Guo
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China,Zhengzhou University, Zhengzhou, China
| | - Peipei Wang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China,Zhengzhou University, Zhengzhou, China
| | - Xuejun Zheng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China,Zhengzhou University, Zhengzhou, China
| | - Wen Cui
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China,Zhengzhou University, Zhengzhou, China
| | - Jin Shang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China,Laboratory of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China,Laboratory Animal Platform of Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China,*Correspondence: Zhanzheng Zhao, ; Jin Shang,
| | - Zhanzheng Zhao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China,Laboratory of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China,Laboratory Animal Platform of Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China,*Correspondence: Zhanzheng Zhao, ; Jin Shang,
| |
Collapse
|
10
|
Wischhof L, Scifo E, Ehninger D, Bano D. AIFM1 beyond cell death: An overview of this OXPHOS-inducing factor in mitochondrial diseases. EBioMedicine 2022; 83:104231. [PMID: 35994922 PMCID: PMC9420475 DOI: 10.1016/j.ebiom.2022.104231] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 11/17/2022] Open
Abstract
Apoptosis-inducing factor (AIF) is a mitochondrial intermembrane space flavoprotein with diverse functions in cellular physiology. In this regard, a large number of studies have elucidated AIF's participation to chromatin condensation during cell death in development, cancer, cardiovascular and brain disorders. However, the discovery of rare AIFM1 mutations in patients has shifted the interest of biomedical researchers towards AIF's contribution to pathogenic mechanisms underlying inherited AIFM1-linked metabolic diseases. The functional characterization of AIF binding partners has rapidly advanced our understanding of AIF biology within the mitochondria and beyond its widely reported role in cell death. At the present time, it is reasonable to assume that AIF contributes to cell survival by promoting biogenesis and maintenance of the mitochondrial oxidative phosphorylation (OXPHOS) system. With this review, we aim to outline the current knowledge around the vital role of AIF by primarily focusing on currently reported human diseases that have been linked to AIFM1 deficiency.
Collapse
Affiliation(s)
- Lena Wischhof
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Enzo Scifo
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Dan Ehninger
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| |
Collapse
|
11
|
Erlandsson MC, Erdogan S, Wasén C, Andersson KME, Silfverswärd ST, Pullerits R, Bemark M, Bokarewa MI. IGF1R signalling is a guardian of self-tolerance restricting autoantibody production. Front Immunol 2022; 13:958206. [PMID: 36105797 PMCID: PMC9464816 DOI: 10.3389/fimmu.2022.958206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/01/2022] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Insulin-like growth factor 1 receptor (IGF1R) acts at the crossroad between immunity and cancer, being an attractive therapeutic target in these areas. IGF1R is broadly expressed by antigen-presenting cells (APC). Using mice immunised with the methylated albumin from bovine serum (BSA-immunised mice) and human CD14+ APCs, we investigated the role that IGF1R plays during adaptive immune responses. METHODS The mBSA-immunised mice were treated with synthetic inhibitor NT157 or short hairpin RNA to inhibit IGF1R signalling, and spleens were analysed by immunohistology and flow cytometry. The levels of autoantibody and cytokine production were measured by microarray or conventional ELISA. The transcriptional profile of CD14+ cells from blood of 55 patients with rheumatoid arthritis (RA) was analysed with RNA-sequencing. RESULTS Inhibition of IGF1R resulted in perifollicular infiltration of functionally compromised S256-phosphorylated FoxO1+ APCs, and an increased frequency of IgM+CD21+ B cells, which enlarged the marginal zone (MZ). Enlargement of MHCII+CD11b+ APCs ensured favourable conditions for their communication with IgM+ B cells in the MZ. The reduced expression of ICOSL and CXCR5 by APCs after IGF1R inhibition led to impaired T cell control, which resulted in autoreactivity of extra-follicular B cells and autoantibody production. In the clinical setting, the low expression of IGF1R on CD14+ APCs was associated with an involuted FOXO pathway, non-inflammatory cell metabolism and a high IL10 production characteristic for tolerogenic macrophages. Furthermore, autoantibody positivity was associated with low IGF1R signalling in CD14+ APCs. CONCLUSIONS In experimental model and in patient material, this study demonstrates that IGF1R plays an important role in preventing autoimmunity. The study raises awareness of that immune tolerance may be broken during therapeutic IGF1R targeting.
Collapse
Affiliation(s)
- Malin C. Erlandsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
- Rheumatology Clinic, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Seval Erdogan
- Department of Rheumatology and Inflammation Research, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Caroline Wasén
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, United States
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Karin M. E. Andersson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Sofia T. Silfverswärd
- Department of Rheumatology and Inflammation Research, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Rille Pullerits
- Department of Rheumatology and Inflammation Research, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mats Bemark
- Department of Clinical Immunology and Transfusion Medicine, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maria I. Bokarewa
- Department of Rheumatology and Inflammation Research, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
- Rheumatology Clinic, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
12
|
Lausecker F, Koehler S, Fresquet M, Naylor RW, Tian P, Wanner N, Braun F, Butt L, Huber TB, Lennon R. Integrating basic science with translational research: the 13th International Podocyte Conference 2021. Kidney Int 2022; 102:708-719. [PMID: 35964799 PMCID: PMC9386279 DOI: 10.1016/j.kint.2022.07.021] [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: 05/12/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 11/30/2022]
Abstract
The 13th International Podocyte Conference was held in Manchester, UK, and online from July 28 to 30, 2021. Originally planned for 2020, this biannual meeting was postponed by a year because of the coronavirus disease 2019 (COVID-19) pandemic and proceeded as an innovative hybrid meeting. In addition to in-person attendance, online registration was offered, and this attracted 490 conference registrations in total. As a Podocyte Conference first, a day for early-career researchers was introduced. This premeeting included talks from graduate students and postdoctoral researchers. It gave early career researchers the opportunity to ask a panel, comprising academic leaders and journal editors, about career pathways and the future for podocyte research. The main meeting over 3 days included a keynote talk and 4 focused sessions each day incorporating invited talks, followed by selected abstract presentations, and an open panel discussion. The conference concluded with a Patient Day, which brought together patients, clinicians, researchers, and industry representatives. The Patient Day was an interactive and diverse day. As well as updates on improving diagnosis and potential new therapies, the Patient Day included a PodoArt competition, exercise and cooking classes with practical nutrition advice, and inspirational stories from patients and family members. This review summarizes the exciting science presented during the 13th International Podocyte Conference and demonstrates the resilience of researchers during a global pandemic.
Collapse
Affiliation(s)
- Franziska Lausecker
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Sybille Koehler
- Biomedical Sciences, University of Edinburgh, Edinburgh, UK; III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maryline Fresquet
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Richard W Naylor
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Pinyuan Tian
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Nicola Wanner
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabian Braun
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Linus Butt
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester University Hospitals National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
13
|
Yang J, Liu Z. Mechanistic Pathogenesis of Endothelial Dysfunction in Diabetic Nephropathy and Retinopathy. Front Endocrinol (Lausanne) 2022; 13:816400. [PMID: 35692405 PMCID: PMC9174994 DOI: 10.3389/fendo.2022.816400] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/28/2022] [Indexed: 12/15/2022] Open
Abstract
Diabetic nephropathy (DN) and diabetic retinopathy (DR) are microvascular complications of diabetes. Microvascular endothelial cells are thought to be the major targets of hyperglycemic injury. In diabetic microvasculature, the intracellular hyperglycemia causes damages to the vascular endothelium, via multiple pathophysiological process consist of inflammation, endothelial cell crosstalk with podocytes/pericytes and exosomes. In addition, DN and DR diseases development are involved in several critical regulators including the cell adhesion molecules (CAMs), the vascular endothelial growth factor (VEGF) family and the Notch signal. The present review attempts to gain a deeper understanding of the pathogenesis complexities underlying the endothelial dysfunction in diabetes diabetic and retinopathy, contributing to the development of new mechanistic therapeutic strategies against diabetes-induced microvascular endothelial dysfunction.
Collapse
Affiliation(s)
- Jing Yang
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zhangsuo Liu
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
14
|
Piazzesi A, Wang Y, Jackson J, Wischhof L, Zeisler-Diehl V, Scifo E, Oganezova I, Hoffmann T, Gómez Martín P, Bertan F, Wrobel CJJ, Schroeder FC, Ehninger D, Händler K, Schultze JL, Schreiber L, van Echten-Deckert G, Nicotera P, Bano D. CEST-2.2 overexpression alters lipid metabolism and extends longevity of mitochondrial mutants. EMBO Rep 2022; 23:e52606. [PMID: 35297148 PMCID: PMC9066074 DOI: 10.15252/embr.202152606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/14/2022] [Accepted: 02/22/2022] [Indexed: 12/17/2022] Open
Abstract
Mitochondrial dysfunction can either extend or decrease Caenorhabditis elegans lifespan, depending on whether transcriptionally regulated responses can elicit durable stress adaptation to otherwise detrimental lesions. Here, we test the hypothesis that enhanced metabolic flexibility is sufficient to circumvent bioenergetic abnormalities associated with the phenotypic threshold effect, thereby transforming short‐lived mitochondrial mutants into long‐lived ones. We find that CEST‐2.2, a carboxylesterase mainly localizes in the intestine, may stimulate the survival of mitochondrial deficient animals. We report that genetic manipulation of cest‐2.2 expression has a minor lifespan impact on wild‐type nematodes, whereas its overexpression markedly extends the lifespan of complex I‐deficient gas‐1(fc21) mutants. We profile the transcriptome and lipidome of cest‐2.2 overexpressing animals and show that CEST‐2.2 stimulates lipid metabolism and fatty acid beta‐oxidation, thereby enhancing mitochondrial respiratory capacity through complex II and LET‐721/ETFDH, despite the inherited genetic lesion of complex I. Together, our findings unveil a metabolic pathway that, through the tissue‐specific mobilization of lipid deposits, may influence the longevity of mitochondrial mutant C. elegans.
Collapse
Affiliation(s)
- Antonia Piazzesi
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Yiru Wang
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Joshua Jackson
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Lena Wischhof
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Enzo Scifo
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Ina Oganezova
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Thorben Hoffmann
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Fabio Bertan
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Chester J J Wrobel
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA
| | - Frank C Schroeder
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA
| | - Dan Ehninger
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Kristian Händler
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE), University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE), University of Bonn, Bonn, Germany.,Department for Genomics and Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| | - Lukas Schreiber
- Institute of Cellular and Molecular Botany (IZMB), University of Bonn, Bonn, Germany
| | | | | | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| |
Collapse
|
15
|
Maremonti F, Meyer C, Linkermann A. Mechanisms and Models of Kidney Tubular Necrosis and Nephron Loss. J Am Soc Nephrol 2022; 33:472-486. [PMID: 35022311 PMCID: PMC8975069 DOI: 10.1681/asn.2021101293] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Understanding nephron loss is a primary strategy for preventing CKD progression. Death of renal tubular cells may occur by apoptosis during developmental and regenerative processes. However, during AKI, the transition of AKI to CKD, sepsis-associated AKI, and kidney transplantation ferroptosis and necroptosis, two pathways associated with the loss of plasma membrane integrity, kill renal cells. This necrotic type of cell death is associated with an inflammatory response, which is referred to as necroinflammation. Importantly, the necroinflammatory response to cells that die by necroptosis may be fundamentally different from the tissue response to ferroptosis. Although mechanisms of ferroptosis and necroptosis have recently been investigated in detail, the cell death propagation during tubular necrosis, although described morphologically, remains incompletely understood. Here, we argue that a molecular switch downstream of tubular necrosis determines nephron regeneration versus nephron loss. Unraveling the details of this "switch" must include the inflammatory response to tubular necrosis and regenerative signals potentially controlled by inflammatory cells, including the stimulation of myofibroblasts as the origin of fibrosis. Understanding in detail the molecular switch and the inflammatory responses to tubular necrosis can inform the discussion of therapeutic options.
Collapse
Affiliation(s)
- Francesca Maremonti
- Division of Nephrology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Claudia Meyer
- Division of Nephrology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Andreas Linkermann
- Division of Nephrology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany .,Biotechnology Center, Technical University of Dresden, Dresden, Germany
| |
Collapse
|
16
|
Audzeyenka I, Bierżyńska A, Lay AC. Podocyte Bioenergetics in the Development of Diabetic Nephropathy: The Role of Mitochondria. Endocrinology 2022; 163:6429716. [PMID: 34791124 PMCID: PMC8660556 DOI: 10.1210/endocr/bqab234] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Indexed: 01/11/2023]
Abstract
Diabetic nephropathy (DN) is the leading cause of kidney failure, with an increasing incidence worldwide. Mitochondrial dysfunction is known to occur in DN and has been implicated in the underlying pathogenesis of disease. These complex organelles have an array of important cellular functions and involvement in signaling pathways, and understanding the intricacies of these responses in health, as well as how they are damaged in disease, is likely to highlight novel therapeutic avenues. A key cell type damaged early in DN is the podocyte, and increasing studies have focused on investigating the role of mitochondria in podocyte injury. This review will summarize what is known about podocyte mitochondrial dynamics in DN, with a particular focus on bioenergetic pathways, highlighting key studies in this field and potential opportunities to target, enhance or protect podocyte mitochondrial function in the treatment of DN.
Collapse
Affiliation(s)
- Irena Audzeyenka
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Gdańsk, Poland
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
- Correspondence: Irena Audzeyenka, PhD, Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Wita Stwosza St. 63, 80-308 Gdansk, Poland.
| | - Agnieszka Bierżyńska
- Bristol Renal, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Abigail C Lay
- Bristol Renal, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
17
|
Guo J, Chiang WC. Mitophagy in aging and longevity. IUBMB Life 2021; 74:296-316. [PMID: 34889504 DOI: 10.1002/iub.2585] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/21/2021] [Indexed: 12/22/2022]
Abstract
The clearance of damaged or unwanted mitochondria by autophagy (also known as mitophagy) is a mitochondrial quality control mechanism postulated to play an essential role in cellular homeostasis, metabolism, and development and confers protection against a wide range of diseases. Proper removal of damaged or unwanted mitochondria is essential for organismal health. Defects in mitophagy are associated with Parkinson's, Alzheimer's disease, cancer, and other degenerative disorders. Mitochondria regulate organismal fitness and longevity via multiple pathways, including cellular senescence, stem cell function, inflammation, mitochondrial unfolded protein response (mtUPR), and bioenergetics. Thus, mitophagy is postulated to be pivotal for maintaining organismal healthspan and lifespan and the protection against aged-related degeneration. In this review, we will summarize recent understanding of the mechanism of mitophagy and aspects of mitochondrial functions. We will focus on mitochondria-related cellular processes that are linked to aging and examine current genetic evidence that supports the hypothesis that mitophagy is a pro-longevity mechanism.
Collapse
Affiliation(s)
- Jing Guo
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Chung Chiang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
18
|
Kim JH, Hwang KH, Dang BTN, Eom M, Kong ID, Gwack Y, Yu S, Gee HY, Birnbaumer L, Park KS, Cha SK. Insulin-activated store-operated Ca 2+ entry via Orai1 induces podocyte actin remodeling and causes proteinuria. Nat Commun 2021; 12:6537. [PMID: 34764278 PMCID: PMC8586150 DOI: 10.1038/s41467-021-26900-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 10/28/2021] [Indexed: 12/28/2022] Open
Abstract
Podocyte, the gatekeeper of the glomerular filtration barrier, is a primary target for growth factor and Ca2+ signaling whose perturbation leads to proteinuria. However, the effects of insulin action on store-operated Ca2+ entry (SOCE) in podocytes remain unknown. Here, we demonstrated that insulin stimulates SOCE by VAMP2-dependent Orai1 trafficking to the plasma membrane. Insulin-activated SOCE triggers actin remodeling and transepithelial albumin leakage via the Ca2+-calcineurin pathway in podocytes. Transgenic Orai1 overexpression in mice causes podocyte fusion and impaired glomerular filtration barrier. Conversely, podocyte-specific Orai1 deletion prevents insulin-stimulated SOCE, synaptopodin depletion, and proteinuria. Podocyte injury and albuminuria coincide with Orai1 upregulation at the hyperinsulinemic stage in diabetic (db/db) mice, which can be ameliorated by the suppression of Orai1-calcineurin signaling. Our results suggest that tightly balanced insulin action targeting podocyte Orai1 is critical for maintaining filter integrity, which provides novel perspectives on therapeutic strategies for proteinuric diseases, including diabetic nephropathy.
Collapse
Affiliation(s)
- Ji-Hee Kim
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Kyu-Hee Hwang
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Bao T N Dang
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Minseob Eom
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - In Deok Kong
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Seyoung Yu
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Heon Yung Gee
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
- Institute of Biomedical Research (BIOMED), School of Medical Sciences, Catholic University of Argentina, C1107AAZ, Buenos Aires, Argentina
| | - Kyu-Sang Park
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Seung-Kuy Cha
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea.
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea.
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea.
| |
Collapse
|
19
|
Lourenço AB, Artal-Sanz M. The Mitochondrial Prohibitin (PHB) Complex in C. elegans Metabolism and Ageing Regulation. Metabolites 2021; 11:metabo11090636. [PMID: 34564452 PMCID: PMC8472356 DOI: 10.3390/metabo11090636] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 12/20/2022] Open
Abstract
The mitochondrial prohibitin (PHB) complex, composed of PHB-1 and PHB-2, is an evolutionarily conserved context-dependent modulator of longevity. This extremely intriguing phenotype has been linked to alterations in mitochondrial function and lipid metabolism. The true biochemical function of the mitochondrial PHB complex remains elusive, but it has been shown to affect membrane lipid composition. Recent work, using large-scale biochemical approaches, has highlighted a broad effect of PHB on the C. elegans metabolic network. Collectively, the biochemical data support the notion that PHB modulates, at least partially, worm longevity through the moderation of fat utilisation and energy production via the mitochondrial respiratory chain. Herein, we review, in a systematic manner, recent biochemical insights into the impact of PHB on the C. elegans metabolome.
Collapse
Affiliation(s)
- Artur B. Lourenço
- Andalusian Centre for Developmental Biology (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, Carretera de Utrera Km 1, 41013 Seville, Spain
- Correspondence: (A.B.L.); (M.A.-S.)
| | - Marta Artal-Sanz
- Andalusian Centre for Developmental Biology (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, Carretera de Utrera Km 1, 41013 Seville, Spain
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Carretera de Utrera Km 1, 41013 Seville, Spain
- Correspondence: (A.B.L.); (M.A.-S.)
| |
Collapse
|
20
|
Lay AC, Hale LJ, Stowell-Connolly H, Pope RJP, Nair V, Ju W, Marquez E, Rollason R, Hurcombe JA, Hayes B, Roberts T, Gillam L, Allington J, Nelson RG, Kretzler M, Holly JMP, Perks CM, McArdle CA, Welsh GI, Coward RJM. IGFBP-1 expression is reduced in human type 2 diabetic glomeruli and modulates β1-integrin/FAK signalling in human podocytes. Diabetologia 2021; 64:1690-1702. [PMID: 33758952 PMCID: PMC8187213 DOI: 10.1007/s00125-021-05427-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 01/14/2021] [Indexed: 12/25/2022]
Abstract
AIMS/HYPOTHESIS Podocyte loss or injury is one of the earliest features observed in the pathogenesis of diabetic kidney disease (DKD), which is the leading cause of end-stage renal failure worldwide. Dysfunction in the IGF axis, including in IGF binding proteins (IGFBPs), is associated with DKD, particularly in the early stages of disease progression. The aim of this study was to investigate the potential roles of IGFBPs in the development of type 2 DKD, focusing on podocytes. METHODS IGFBP expression was analysed in the Pima DKD cohort, alongside data from the Nephroseq database, and in ex vivo human glomeruli. Conditionally immortalised human podocytes and glomerular endothelial cells were studied in vitro, where IGFBP-1 expression was analysed using quantitative PCR and ELISAs. Cell responses to IGFBPs were investigated using migration, cell survival and adhesion assays; electrical cell-substrate impedance sensing; western blotting; and high-content automated imaging. RESULTS Data from the Pima DKD cohort and from the Nephroseq database demonstrated a significant reduction in glomerular IGFBP-1 in the early stages of human type 2 DKD. In the glomerulus, IGFBP-1 was predominantly expressed in podocytes and controlled by phosphoinositide 3-kinase (PI3K)-forkhead box O1 (FoxO1) activity. In vitro, IGFBP-1 signalled to podocytes via β1-integrins, resulting in increased phosphorylation of focal-adhesion kinase (FAK), increasing podocyte motility, adhesion, electrical resistance across the adhesive cell layer and cell viability. CONCLUSIONS/INTERPRETATION This work identifies a novel role for IGFBP-1 in the regulation of podocyte function and that the glomerular expression of IGFBP-1 is reduced in the early stages of type 2 DKD, via reduced FoxO1 activity. Thus, we hypothesise that strategies to maintain glomerular IGFBP-1 levels may be beneficial in maintaining podocyte function early in DKD.
Collapse
Affiliation(s)
- Abigail C Lay
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Lorna J Hale
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | | | - Robert J P Pope
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Viji Nair
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Wenjun Ju
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Eva Marquez
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ruth Rollason
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Jenny A Hurcombe
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Bryony Hayes
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Timothy Roberts
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Lawrence Gillam
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Jonathan Allington
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Robert G Nelson
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Jeff M P Holly
- IGFs and Metabolic Endocrinology Group, Bristol Medical School, University of Bristol, Bristol, UK
| | - Claire M Perks
- IGFs and Metabolic Endocrinology Group, Bristol Medical School, University of Bristol, Bristol, UK
| | - Craig A McArdle
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Gavin I Welsh
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Richard J M Coward
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK.
| |
Collapse
|
21
|
Liu L, Ma F, Hao Y, Yi Z, Yu X, Xu B, Wei C, Hu J. Integrative Informatics Analysis of Transcriptome and Identification of Interacted Genes in the Glomeruli and Tubules in CKD. Front Med (Lausanne) 2021; 7:615306. [PMID: 33644086 PMCID: PMC7906987 DOI: 10.3389/fmed.2020.615306] [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: 10/08/2020] [Accepted: 12/23/2020] [Indexed: 11/16/2022] Open
Abstract
Chronic kidney disease (CKD) is a complex disease in which the renal function is compromised chronically. Many studies have indicated the crosstalk between the tubule and the glomerulus in CKD progression. However, our understanding of the interaction of tubular and glomerular injury remains incomplete. In this study, we applied a meta-analysis approach on the transcriptome of the tubules and glomeruli of CKD patients to identify differentially expressed genes (DEGs) signature. Functional analysis of pathways and Gene Ontology found that tubular DEGs were mainly involved in cell assembly and remodeling, glomerular DEGs in cell proliferation and apoptosis, and overlapping DEGs mainly in immune response. Correlation analysis was performed to identify the associated DEGs in the tubules and glomeruli. Secreted protein comparison and verification experiments indicated that WFDC2 from the tubule could downregulate PEX19 mRNA and protein levels at the glomeruli in diabetic kidney disease (DKD). This study revealed the distinctive pathways of the tubules and glomeruli and identified interacted genes during CKD progression.
Collapse
Affiliation(s)
- Lingyun Liu
- Department of Andrology, The First Hospital of Jilin University, Jilin, China
| | - Fuzhe Ma
- Department of Nephrology, The First Hospital of Jilin University, Jilin, China
| | - Yuanyuan Hao
- Department of Urology, The First Hospital of Jilin University, Jilin, China
| | - Zhengzi Yi
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Xiaoxia Yu
- Division of Nephrology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Bo Xu
- Department of Urology, The First Hospital of Jilin University, Jilin, China
| | - Chengguo Wei
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jinghai Hu
- Department of Urology, The First Hospital of Jilin University, Jilin, China
| |
Collapse
|
22
|
Son SS, Kang JS, Lee EY. Paclitaxel Ameliorates Palmitate-Induced Injury in Mouse Podocytes. Med Sci Monit Basic Res 2020; 26:e928265. [PMID: 33323915 PMCID: PMC7751256 DOI: 10.12659/msmbr.928265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background Palmitate, a common saturated free fatty acid, is increased in patients with diabetic nephropathy (DN). Excessive palmitate in kidney is known to cause proteinuria and fibrosis. Several studies have demonstrated that paclitaxel has anti-fibrotic and anti-inflammatory effects on kidney disease. However, whether paclitaxel can relieve podocyte injury is unclear. Material/Methods Immortalized mouse podocytes were used as an in vitro system. Palmitate was used to induce podocyte injury. Podocytes were divided into 4 groups: bovine serum albumin, palmitate, palmitate+1 nM paclitaxel, and palmitate+5 nM paclitaxel. The effects of paclitaxel on palmitate-induced podocyte injury were analyzed by western blot and real-time PCR. Intracellular reactive oxygen species (ROS) generation and podocyte cytoskeletons were analyzed using CM-H2DCF-DA and phalloidin staining. Results Paclitaxel restored downregulated expression of nephrin and synaptopodin and upregulated VEGF expression after injury induced by palmitate. Remarkably, palmitate-induced actin cytoskeleton rearrangement in podocytes was repaired by paclitaxel. Four endoplasmic reticulum stress markers, ATF-6α, Bip, CHOP, and spliced xBP1, were significantly increased in palmitate-treated podocytes compared with control podocytes. Such increases were decreased by paclitaxel treatment. Palmitate-induced ROS generation was ameliorated by paclitaxel. Elevated Nox4 expression was also improved by paclitaxel. Paclitaxel alleviated the expression levels of the antioxidant molecules, Nrf-2, HO-1, SOD-1, and SOD-2. The paclitaxel effects were accompanied by inhibition of the inflammatory cytokines, MCP-1, TNF-α, TNF-R2, and TLR4, as well as attenuation of the apoptosis markers, Bax, Bcl-2, and Caspase-3. Furthermore, paclitaxel suppressed the palmitate-induced fibrosis molecules, fibronectin and TGF-β1. Conclusions This study suggests that paclitaxel could be a therapeutic agent for treating palmitate-induced podocyte injury in DN.
Collapse
Affiliation(s)
- Seung Seob Son
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, South Korea
| | - Jeong Suk Kang
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, South Korea.,Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan, South Korea
| | - Eun Young Lee
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, South Korea.,Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan, South Korea.,Department of Internal Medicine, BK21 Four Project, College of Medicine, Soonchunhyang University, Cheonan, South Korea
| |
Collapse
|
23
|
Abstract
The current paradigm of type 2 diabetes (T2D) is gluco-centric, being exclusively categorized by glycemic characteristics. The gluco-centric paradigm views hyperglycemia as the primary target, being driven by resistance to insulin combined with progressive beta cells failure, and considers glycemic control its ultimate treatment goal. Most importantly, the gluco-centric paradigm considers the non-glycemic diseases associated with T2D, e.g., obesity, dyslipidemia, hypertension, macrovascular disease, microvascular disease and fatty liver as 'risk factors' and/or 'outcomes' and/or 'comorbidities', rather than primary inherent disease aspects of T2D. That is in spite of their high prevalence (60-90%) and major role in profiling T2D morbidity and mortality. Moreover, the gluco-centric paradigm fails to realize that the non-glycemic diseases of T2D are driven by insulin and, except for glycemic control, response to insulin in T2D is essentially the rule rather than the exception. Failure of the gluco-centric paradigm to offer an exhaustive unifying view of the glycemic and non-glycemic diseases of T2D may have contributed to T2D being still an unmet need. An mTORC1-centric paradigm maintains that hyperactive mTORC1 drives the glycemic and non-glycemic disease aspects of T2D. Hyperactive mTORC1 is proposed to act as double-edged agent, namely, to interfere with glycemic control by disrupting the insulin receptor-Akt transduction pathway, while concomitantly driving the non-glycemic diseases of T2D. The mTORC1-centric paradigm may offer a novel perspective for T2D in terms of pathogenesis, clinical focus and treatment strategy.
Collapse
Affiliation(s)
- Jacob Bar-Tana
- Hebrew University Medical School, 91120, Jerusalem, Israel.
| |
Collapse
|
24
|
Martin-Perez M, Grillo AS, Ito TK, Valente AS, Han J, Entwisle SW, Huang HZ, Kim D, Yajima M, Kaeberlein M, Villén J. PKC downregulation upon rapamycin treatment attenuates mitochondrial disease. Nat Metab 2020; 2:1472-1481. [PMID: 33324011 PMCID: PMC8017771 DOI: 10.1038/s42255-020-00319-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/06/2020] [Indexed: 02/06/2023]
Abstract
Leigh syndrome is a fatal neurometabolic disorder caused by defects in mitochondrial function. Mechanistic target of rapamycin (mTOR) inhibition with rapamycin attenuates disease progression in a mouse model of Leigh syndrome (Ndufs4 knock-out (KO) mouse); however, the mechanism of rescue is unknown. Here we identify protein kinase C (PKC) downregulation as a key event mediating the beneficial effects of rapamycin treatment of Ndufs4 KO mice. Assessing the impact of rapamycin on the brain proteome and phosphoproteome of Ndufs4 KO mice, we find that rapamycin restores mitochondrial protein levels, inhibits signalling through both mTOR complexes and reduces the abundance and activity of multiple PKC isoforms. Administration of PKC inhibitors increases survival, delays neurological deficits, prevents hair loss and decreases inflammation in Ndufs4 KO mice. Thus, PKC may be a viable therapeutic target for treating severe mitochondrial disease.
Collapse
Affiliation(s)
- Miguel Martin-Perez
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, Barcelona, Spain
| | - Anthony S Grillo
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Takashi K Ito
- Department of Pathology, University of Washington, Seattle, WA, USA
- RIKEN Center for Sustainable Resource Science, Saitama, Japan
| | - Anthony S Valente
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Jeehae Han
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Samuel W Entwisle
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Heather Z Huang
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Dayae Kim
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Masanao Yajima
- Department of Mathematics and Statistics, Boston University, Boston, MA, USA
| | - Matt Kaeberlein
- Department of Pathology, University of Washington, Seattle, WA, USA.
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, USA.
| | - Judit Villén
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, USA.
| |
Collapse
|
25
|
Burek M, Burmester S, Salvador E, Möller-Ehrlich K, Schneider R, Roewer N, Nagai M, Förster CY. Kidney Ischemia/Reperfusion Injury Induces Changes in the Drug Transporter Expression at the Blood-Brain Barrier in vivo and in vitro. Front Physiol 2020; 11:569881. [PMID: 33281613 PMCID: PMC7688901 DOI: 10.3389/fphys.2020.569881] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 10/20/2020] [Indexed: 01/25/2023] Open
Abstract
Ischemia/reperfusion injury is a major cause of acute kidney injury (AKI). AKI is characterized by a sudden decrease in kidney function, systemic inflammation, oxidative stress, and dysregulation of the sodium, potassium, and water channels. While AKI leads to uremic encephalopathy, epidemiological studies have shown that AKI is associated with a subsequent risk for developing stroke and dementia. To get more insights into kidney-brain crosstalk, we have created an in vitro co-culture model based on human kidney cells of the proximal tubule (HK-2) and brain microvascular endothelial cells (BMEC). The HK-2 cell line was grown to confluence on 6-well plates and exposed to oxygen/glucose deprivation (OGD) for 4 h. Control HK-2 cells were grown under normal conditions. The BMEC cell line cerebED was grown to confluence on transwells with 0.4 μm pores. The transwell filters seeded and grown to confluence with cereEND were inserted into the plates with HK-2 cells with or without OGD treatment. In addition, cerebEND were left untreated or treated with uremic toxins, indole-3-acetic acid (IAA) and indoxyl sulfate (IS). The protein and mRNA expression of selected BBB-typical influx transporters, efflux transporters, cellular receptors, and tight junction proteins was measured in BMECs. To validate this in vitro model of kidney-brain interaction, we isolated brain capillaries from mice exposed to bilateral renal ischemia (30 min)/reperfusion injury (24 h) and measured mRNA and protein expression as described above. Both in vitro and in vivo systems showed similar changes in the expression of drug transporters, cellular receptors, and tight junction proteins. Efflux pumps, in particular Abcb1b, Abcc1, and Abcg2, have shown increased expression in our model. Thus, our in vitro co-culture system can be used to study the cellular mechanism of kidney and brain crosstalk in renal ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Malgorzata Burek
- Department of Anaesthesia and Critical Care, University of Würzburg, Würzburg, Germany
| | - Sandra Burmester
- Department of Anaesthesia and Critical Care, University of Würzburg, Würzburg, Germany
| | - Ellaine Salvador
- Department of Anaesthesia and Critical Care, University of Würzburg, Würzburg, Germany
| | - Kerstin Möller-Ehrlich
- Division of Nephrology, Department of Medicine I, University of Würzburg, Würzburg, Germany
| | - Reinhard Schneider
- Division of Nephrology, Department of Medicine I, University of Würzburg, Würzburg, Germany
| | - Norbert Roewer
- Department of Anaesthesia and Critical Care, University of Würzburg, Würzburg, Germany
| | - Michiaki Nagai
- Department of Cardiology, Hiroshima City Asa Hospital, Hiroshima, Japan
| | - Carola Y. Förster
- Department of Anaesthesia and Critical Care, University of Würzburg, Würzburg, Germany
| |
Collapse
|
26
|
Mechanism of progression of diabetic kidney disease mediated by podocyte mitochondrial injury. Mol Biol Rep 2020; 47:8023-8035. [PMID: 32918716 DOI: 10.1007/s11033-020-05749-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 08/28/2020] [Indexed: 12/21/2022]
Abstract
Diabetic kidney disease (DKD) is an important diabetic microvascular complication, which has become the main cause of end-stage renal disease (ESRD) all over the world. It is of great significance to find effective therapeutic targets and improve the prognosis of the disease. Traditionally, it is believed that the activation of the renin-angiotensin-aldosterone system (RAAS) is the main reason for the progression of DKD, but with the progress of research, it is known that the production of proteinuria in patients with DKD is also related to podocyte injury and loss. Many studies have shown that mitochondrial dysfunction in podocytes plays an important role in the occurrence and development of DKD, and oxidative stress is also the main pathway and common hub of diabetes to the occurrence and development of microvascular and macrovascular complications. Thus, the occurrence and progression of DKD is correlated with not only the activation of the RAAS, but also the damage of mitochondria, oxidative stress, and inflammatory mediators. Besides, diabetes-related metabolic disorders can also cause abnormalities in mitochondrial dynamics, autophagy and cellular signal transduction, which are intertwined in a complex way. Therefore, in this review, we mainly explore the mechanism and the latest research progress of podocyte mitochondria in DKD and summarize the main signal pathways involved in them. Thus, it provides feasible clinical application and future research suggestions for the prevention and treatment of DKD, which has important practical significance for the later treatment of patients with DKD.
Collapse
|
27
|
Shi Y, Li Q, Sun F, Zhu C, Ma S, Qin D, Li Q, Li T. Lamprey PHB2 maintains mitochondrial stability by tanslocation to the mitochondria under oxidative stress. FISH & SHELLFISH IMMUNOLOGY 2020; 104:613-621. [PMID: 32592929 PMCID: PMC7311904 DOI: 10.1016/j.fsi.2020.06.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 06/11/2023]
Abstract
Before we have reported lamprey PHB2 could enhance the cellular oxidative-stressed tolerance, here the aim was to explore its mechanisms. We used flow cytometry analysis to identify a Lampetra morii homologue of PHB2 (Lm-PHB2) that could significantly decrease the levels of ROS generation in HEK293T cells. According to confocal microscopy observations, Lm-PHB2 contributed to maintain the mitochondrial morphology of HEK293T cells, and then both cellular nuclear location and translocation from the nucleus to mitochondria of Lm-PHB2 were also examined in HEK293T cells under oxidative stress. We also examined the expressions and locations of various Lm-PHB2 deletion mutants and the amino acid mutant by confocal microscopy and the results showed that the translocation of Lm-PHB2 into mitochondria was dependent on the Lm-PHB21-50aa region and the 17th, 48th and 57th three arginines (R) of N-terminal were very critical. In addition, the analyses of QRT-PCR and Western blot demonstrated that Lm-PHB2 increased the expression levels of OPA1 and HAX1 in HEK293T cells treated with H2O2. The analyses of immunofluorescence and immunoprecipitation showed that Lm-PHB2 could interact with OPA1 and HAX1, respectively. The above mentioned results indicate that Lm-PHB2 could assist OPA1 and HAX1 to maintain mitochondrial morphology and decrease ROS levels by the translocation from the nucleus to mitochondria under oxidative stress.
Collapse
Affiliation(s)
- Ying Shi
- College of Life Sciences, Lamprey Research Center, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, 116081, China
| | - Qing Li
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, 110169, China
| | - Feng Sun
- College of Life Sciences, Lamprey Research Center, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, 116081, China; Collaborative Innovation Center of Marine Food Deep Processing, Dalian Polytechnic University, Dalian, 116034, China
| | - Chenyue Zhu
- College of Life Sciences, Lamprey Research Center, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, 116081, China
| | - Sainan Ma
- College of Life Sciences, Lamprey Research Center, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, 116081, China
| | - Di Qin
- College of Life Sciences, Lamprey Research Center, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, 116081, China
| | - Qingwei Li
- College of Life Sciences, Lamprey Research Center, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, 116081, China; Collaborative Innovation Center of Marine Food Deep Processing, Dalian Polytechnic University, Dalian, 116034, China.
| | - Tiesong Li
- College of Life Sciences, Lamprey Research Center, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, 116081, China; Collaborative Innovation Center of Marine Food Deep Processing, Dalian Polytechnic University, Dalian, 116034, China.
| |
Collapse
|
28
|
Abstract
Neuropeptide Y (NPY) is implicated in many pathological conditions including obesity, diabetes, and insulin resistance. However, a pathogenic role of NPY in kidney disease has not been described. We found that NPY is produced by the podocyte in the glomerulus, and this production decreases in renal disease, in contrast to an increase in circulating NPY levels. In the glomerulus, NPY signals via the NPY receptor 2 (NPY2R) and modulates PI3K, MAPK, and NFAT signaling, along with RNA processing and cell migration and, if prolonged, predicted nephrotoxicity. The pharmacological inhibition of NPY-NPY2R signaling also protected against albuminuria and kidney disease in a mouse model of glomerulosclerosis, suggesting that inhibiting this pathway may be therapeutically beneficial in the prevention of kidney disease. Albuminuria is an independent risk factor for the progression to end-stage kidney failure, cardiovascular morbidity, and premature death. As such, discovering signaling pathways that modulate albuminuria is desirable. Here, we studied the transcriptomes of podocytes, key cells in the prevention of albuminuria, under diabetic conditions. We found that Neuropeptide Y (NPY) was significantly down-regulated in insulin-resistant vs. insulin-sensitive mouse podocytes and in human glomeruli of patients with early and late-stage diabetic nephropathy, as well as other nondiabetic glomerular diseases. This contrasts with the increased plasma and urinary levels of NPY that are observed in such conditions. Studying NPY-knockout mice, we found that NPY deficiency in vivo surprisingly reduced the level of albuminuria and podocyte injury in models of both diabetic and nondiabetic kidney disease. In vitro, podocyte NPY signaling occurred via the NPY2 receptor (NPY2R), stimulating PI3K, MAPK, and NFAT activation. Additional unbiased proteomic analysis revealed that glomerular NPY-NPY2R signaling predicted nephrotoxicity, modulated RNA processing, and inhibited cell migration. Furthermore, pharmacologically inhibiting the NPY2R in vivo significantly reduced albuminuria in adriamycin-treated glomerulosclerotic mice. Our findings suggest a pathogenic role of excessive NPY-NPY2R signaling in the glomerulus and that inhibiting NPY-NPY2R signaling in albuminuric kidney disease has therapeutic potential.
Collapse
|
29
|
Chen S, Lv L, Liu B, Tang R. Crosstalk between tubular epithelial cells and glomerular endothelial cells in diabetic kidney disease. Cell Prolif 2020; 53:e12763. [PMID: 31925859 PMCID: PMC7106959 DOI: 10.1111/cpr.12763] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/26/2019] [Accepted: 12/21/2019] [Indexed: 12/21/2022] Open
Abstract
In recent years, although the development of clinical therapy for diabetic kidney disease (DKD) has made great progress, the progression of DKD still cannot be controlled. Therefore, further study of the pathogenesis of DKD and improvements in DKD treatment are crucial for prognosis. Traditional studies have shown that podocyte injury plays an important role in this process. Recently, it has been found that glomerulotubular balance and tubuloglomerular feedback (TGF) may be involved in the progression of DKD. Glomerulotubular balance is the specific gravity absorption of the glomerular ultrafiltrate by the proximal tubules, which absorbs only 65% to 70% of the ultrafiltrate. This ensures that the urine volume will not change much regardless of whether the glomerular filtration rate (GFR) increases or decreases. TGF is one of the significant mechanisms of renal blood flow and self-regulation of GFR, but how they participate in the development of DKD in the pathological state and the specific mechanism is not clear. Injury to tubular epithelial cells (TECs) is the key link in DKD. Additionally, injury to glomerular endothelial cells (GECs) plays a key role in the early occurrence and development of DKD. However, TECs and GECs are close to each other in anatomical position and can crosstalk with each other, which may affect the development of DKD. Therefore, the purpose of this review was to summarize the current knowledge on the crosstalk between TECs and GECs in the pathogenesis of DKD and to highlight specific clinical and potential therapeutic strategies.
Collapse
Affiliation(s)
- Si‐Jie Chen
- Institute of NephrologyZhongda HospitalNanjing Lishui People's HospitalNanjingChina
- Institute of NephrologyZhongda HospitalSchool of MedicineSoutheast UniversityNanjingChina
| | - Lin‐Li Lv
- Institute of NephrologyZhongda HospitalSchool of MedicineSoutheast UniversityNanjingChina
| | - Bi‐Cheng Liu
- Institute of NephrologyZhongda HospitalNanjing Lishui People's HospitalNanjingChina
- Institute of NephrologyZhongda HospitalSchool of MedicineSoutheast UniversityNanjingChina
| | - Ri‐Ning Tang
- Institute of NephrologyZhongda HospitalNanjing Lishui People's HospitalNanjingChina
- Institute of NephrologyZhongda HospitalSchool of MedicineSoutheast UniversityNanjingChina
| |
Collapse
|
30
|
Lehtonen S. SHIPping out diabetes-Metformin, an old friend among new SHIP2 inhibitors. Acta Physiol (Oxf) 2020; 228:e13349. [PMID: 31342643 PMCID: PMC6916339 DOI: 10.1111/apha.13349] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 07/15/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023]
Abstract
SHIP2 (Src homology 2 domain‐containing inositol 5′‐phosphatase 2) belongs to the family of 5′‐phosphatases. It regulates the phosphoinositide 3‐kinase (PI3K)‐mediated insulin signalling cascade by dephosphorylating the 5′‐position of PtdIns(3,4,5)P3 to generate PtdIns(3,4)P2, suppressing the activity of the pathway. SHIP2 mouse models and genetic studies in human propose that increased expression or activity of SHIP2 contributes to the pathogenesis of the metabolic syndrome, hypertension and type 2 diabetes. This has raised great interest to identify SHIP2 inhibitors that could be used to design new treatments for metabolic diseases. This review summarizes the central mechanisms associated with the development of diabetic kidney disease, including the role of insulin resistance, and then moves on to describe the function of SHIP2 as a regulator of metabolism in mouse models. Finally, the identification of SHIP2 inhibitors and their effects on metabolic processes in vitro and in vivo are outlined. One of the newly identified SHIP2 inhibitors is metformin, the first‐line medication prescribed to patients with type 2 diabetes, further boosting the attraction of SHIP2 as a treatment target to ameliorate metabolic disorders.
Collapse
Affiliation(s)
- Sanna Lehtonen
- Department of Pathology and Research Program for Clinical and Molecular Metabolism, Faculty of Medicine University of Helsinki Helsinki Finland
| |
Collapse
|
31
|
Wang J, Zhu P, Li R, Ren J, Zhang Y, Zhou H. Bax inhibitor 1 preserves mitochondrial homeostasis in acute kidney injury through promoting mitochondrial retention of PHB2. Theranostics 2020; 10:384-397. [PMID: 31903127 PMCID: PMC6929616 DOI: 10.7150/thno.40098] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 09/13/2019] [Indexed: 12/21/2022] Open
Abstract
Bax inhibitor-1 (BI1) conveys anti-apoptotic signals for mitochondria while prohibitin 2 (PHB2) is implicated in sustaining mitochondrial morphology and function. However, their regulatory roles in acute kidney injury (AKI) are largely unknown. Methods: In human patients with AKI, levels of BI1 in urine and plasma were determined using ELISA. An experimental model of AKI was established using ATP depletion-mediated metabolic stress and ischemia-reperfusion injury (IRI) in primary tubule cells and BI1 transgenic mice, respectively. Western blots, ELISA, qPCR, immunofluorescence, RNA silencing, and domain deletion assay were employed to evaluate the roles of BI1 and PHB2 in the preservation of mitochondrial integrity. Results: Levels of BI1 in urine and plasma were decreased in patients with AKI and its expression correlated inversely with renal function. However, reconstitution of BI1 in a murine AKI model was capable of alleviating renal failure, inflammation and tubular death. Further molecular scrutiny revealed that BI1 preserved mitochondrial genetic integrity, reduced mitochondrial oxidative stress, promoted mitochondrial respiration, inhibited excessive mitochondrial fission, improved mitophagy and suppressed mitochondrial apoptosis. Intriguingly, levels of the mitochondria-localized PHB2 were sustained by BI1 and knockdown of PHB2 abolished the mitochondrial- and renal- protective properties of BI1. Furthermore, BI1 promoted PHB2 retention within mitochondria through direct interaction with cytoplasmic PHB2 to facilitate its mitochondrial import. This was confirmed by the observation that the C-terminus of BI1 and the PHB domain of PHB2 were required for the BI1-PHB2 cross-linking. Conclusion: Our data have unveiled an essential role of BI1 as a master regulator of renal tubule function through sustaining mitochondrial localization of PHB2, revealing novel therapeutic promises against AKI.
Collapse
|
32
|
Wu HH, Chen CJ, Lin PY, Liu YH. Involvement of prohibitin 1 and prohibitin 2 upregulation in cBSA-induced podocyte cytotoxicity. J Food Drug Anal 2019; 28:183-194. [PMID: 31883607 DOI: 10.1016/j.jfda.2019.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/15/2019] [Accepted: 09/20/2019] [Indexed: 10/25/2022] Open
Abstract
Membranous nephropathy (MN) is the most common cause of nephrotic syndrome in adults, when not effectively treated. The aim of this study was to discover new targets for the diagnosis and treatment of MN. A reliable mouse model of MN was used by the administration of cationic bovine serum albumin (cBSA). Mice with MN exhibited proteinuria, histopathological changes, and accumulation of immune complexes in the glomerular basement membrane. Label-free proteomics analysis was performed to identify changes in protein expression, and the overexpressed proteins were evaluated. There were 273 proteins that showed significantly different expression in mice with MN, as compared to the controls. String analysis showed that functions related to cellular catabolic processes were downregulated in MN. Among the differentially expressed proteins, prohibitin 1 (PHB1) and prohibitin 2 (PHB2) were upregulated in the kidneys of mice with MN, as demonstrated by immunohistochemistry (IHC), and this upregulation was observed in both the tubular cells and glomeruli. Both shRNA-mediated knockdown of PHB1 or PHB2 inhibited tumor suppressor p53 expression and significantly promoted podocyte proliferation. In addition, both PHB1 and PHB2 were responsible for cBSA-induced cytotoxicity. Microarray analysis further revealed that the upregulation of PHB1 and PHB2 may be due to a blockage of proteasome activity. These data demonstrate that the upregulation of PHB2 is involved in cBSA-mediated podocyte cytotoxicity, which may lead to MN development.
Collapse
Affiliation(s)
- Heng-Hsiung Wu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Chao-Jung Chen
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Pei-Yu Lin
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan; Department of Medical Genetics and Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Yu-Huei Liu
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan; Department of Medical Genetics and Medical Research, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
33
|
Andrade MJ, Van Lonkhuyzen DR, Upton Z, Satyamoorthy K. Unravelling the insulin-like growth factor I-mediated photoprotection of the skin. Cytokine Growth Factor Rev 2019; 52:45-55. [PMID: 31767341 DOI: 10.1016/j.cytogfr.2019.11.004] [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: 10/17/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 10/25/2022]
Abstract
Chronic exposure of human skin to solar ultraviolet radiation (UVR) induces a range of biological reactions which may directly or indirectly lead to the development of skin cancer. In order to overcome these damaging effects of UVR and to reduce photodamage, the skin's endogenous defence system functions in concert with the various exogenous photoprotectors. Growth factors, particularly insulin-like growth factor-I (IGF-I), produced within the body as a result of cellular interaction in response to UVR demonstrates photoprotective properties in human skin. This review summarises the impact of UVR-induced photolesions on human skin, discusses various endogenous as well as exogenous approaches of photoprotection described to date and explains how IGF-I mediates UVR photoprotective responses at the cellular and mitochondrial level. Further, we describe the current interventions using growth factors and propose how the knowledge of the IGF-I photoprotection signalling cascades may direct the development of improved UVR protection and remedial strategies.
Collapse
Affiliation(s)
- Melisa J Andrade
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India; Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Derek R Van Lonkhuyzen
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Zee Upton
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia; Institute of Medical Biology, A⁎STAR, Singapore
| | - Kapaettu Satyamoorthy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
34
|
Torban E, Braun F, Wanner N, Takano T, Goodyer PR, Lennon R, Ronco P, Cybulsky AV, Huber TB. From podocyte biology to novel cures for glomerular disease. Kidney Int 2019; 96:850-861. [PMID: 31420194 DOI: 10.1016/j.kint.2019.05.015] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/23/2019] [Accepted: 05/13/2019] [Indexed: 01/20/2023]
Abstract
The podocyte is a key component of the glomerular filtration barrier. Podocyte dysfunction is central to the underlying pathophysiology of many common glomerular diseases, including diabetic nephropathy, glomerulonephritis and genetic forms of nephrotic syndrome. Collectively, these conditions affect millions of people worldwide, and account for the majority of kidney diseases requiring dialysis and transplantation. The 12th International Podocyte Conference was held in Montreal, Canada from May 30 to June 2, 2018. The primary aim of this conference was to bring together nephrologists, clinician scientists, basic scientists and their trainees from all over the world to present their research and to establish networks with the common goal of developing new therapies for glomerular diseases based on the latest advances in podocyte biology. This review briefly highlights recent advances made in understanding podocyte structure and metabolism, experimental systems in which to study podocytes and glomerular disease, disease mediators, genetic and immune origins of glomerulopathies, and the development of novel therapeutic agents to protect podocyte and glomerular injury.
Collapse
Affiliation(s)
- Elena Torban
- Department of Medicine, McGill University Health Centre Research Institute, McGill University, Montreal, Quebec, Canada.
| | - Fabian Braun
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Wanner
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tomoko Takano
- Department of Medicine, McGill University Health Centre Research Institute, McGill University, Montreal, Quebec, Canada
| | - Paul R Goodyer
- Department of Pediatrics, McGill University Health Centre Research Institute, McGill University, Montreal, Quebec, Canada
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Pierre Ronco
- Sorbonne University, INSERM UMR_S 1155, and Nephrology and Dialysis Department, Hôpital Tenon, Paris France
| | - Andrey V Cybulsky
- Department of Medicine, McGill University Health Centre Research Institute, McGill University, Montreal, Quebec, Canada
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
35
|
Brinkkoetter PT, Bork T, Salou S, Liang W, Mizi A, Özel C, Koehler S, Hagmann HH, Ising C, Kuczkowski A, Schnyder S, Abed A, Schermer B, Benzing T, Kretz O, Puelles VG, Lagies S, Schlimpert M, Kammerer B, Handschin C, Schell C, Huber TB. Anaerobic Glycolysis Maintains the Glomerular Filtration Barrier Independent of Mitochondrial Metabolism and Dynamics. Cell Rep 2019; 27:1551-1566.e5. [PMID: 31042480 PMCID: PMC6506687 DOI: 10.1016/j.celrep.2019.04.012] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 03/13/2019] [Accepted: 04/02/2019] [Indexed: 12/22/2022] Open
Abstract
The cellular responses induced by mitochondrial dysfunction remain elusive. Intrigued by the lack of almost any glomerular phenotype in patients with profound renal ischemia, we comprehensively investigated the primary sources of energy of glomerular podocytes. Combining functional measurements of oxygen consumption rates, glomerular metabolite analysis, and determination of mitochondrial density of podocytes in vivo, we demonstrate that anaerobic glycolysis and fermentation of glucose to lactate represent the key energy source of podocytes. Under physiological conditions, we could detect neither a developmental nor late-onset pathological phenotype in podocytes with impaired mitochondrial biogenesis machinery, defective mitochondrial fusion-fission apparatus, or reduced mtDNA stability and transcription caused by podocyte-specific deletion of Pgc-1α, Drp1, or Tfam, respectively. Anaerobic glycolysis represents the predominant metabolic pathway of podocytes. These findings offer a strategy to therapeutically interfere with the enhanced podocyte metabolism in various progressive kidney diseases, such as diabetic nephropathy or focal segmental glomerulosclerosis (FSGS).
Collapse
Affiliation(s)
- Paul T Brinkkoetter
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Cologne, Germany
| | - Tillmann Bork
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sarah Salou
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wei Liang
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Athanasia Mizi
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Cologne, Germany
| | - Cem Özel
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Cologne, Germany
| | - Sybille Koehler
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Cologne, Germany
| | - H Henning Hagmann
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Cologne, Germany
| | - Christina Ising
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Cologne, Germany
| | - Alexander Kuczkowski
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Cologne, Germany
| | | | - Ahmed Abed
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Cologne, Germany
| | - Oliver Kretz
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Victor G Puelles
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Division of Nephrology and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany; Department of Nephrology, Monash Health, Melbourne, VIC, Australia
| | - Simon Lagies
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Manuel Schlimpert
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Bernd Kammerer
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | | | - Christoph Schell
- Institute of Surgical Pathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
36
|
Gioran A, Piazzesi A, Bertan F, Schroer J, Wischhof L, Nicotera P, Bano D. Multi-omics identify xanthine as a pro-survival metabolite for nematodes with mitochondrial dysfunction. EMBO J 2019; 38:embj.201899558. [PMID: 30796049 PMCID: PMC6418696 DOI: 10.15252/embj.201899558] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 12/10/2018] [Accepted: 01/18/2019] [Indexed: 12/13/2022] Open
Abstract
Aberrant mitochondrial function contributes to the pathogenesis of various metabolic and chronic disorders. Inhibition of insulin/IGF‐1 signaling (IIS) represents a promising avenue for the treatment of mitochondrial diseases, although many of the molecular mechanisms underlying this beneficial effect remain elusive. Using an unbiased multi‐omics approach, we report here that IIS inhibition reduces protein synthesis and favors catabolism in mitochondrial deficient Caenorhabditis elegans. We unveil that the lifespan extension does not occur through the restoration of mitochondrial respiration, but as a consequence of an ATP‐saving metabolic rewiring that is associated with an evolutionarily conserved phosphoproteome landscape. Furthermore, we identify xanthine accumulation as a prominent downstream metabolic output of IIS inhibition. We provide evidence that supplementation of FDA‐approved xanthine derivatives is sufficient to promote fitness and survival of nematodes carrying mitochondrial lesions. Together, our data describe previously unknown molecular components of a metabolic network that can extend the lifespan of short‐lived mitochondrial mutant animals.
Collapse
Affiliation(s)
- Anna Gioran
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Antonia Piazzesi
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Fabio Bertan
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Jonas Schroer
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Lena Wischhof
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| |
Collapse
|
37
|
Signorile A, Sgaramella G, Bellomo F, De Rasmo D. Prohibitins: A Critical Role in Mitochondrial Functions and Implication in Diseases. Cells 2019; 8:cells8010071. [PMID: 30669391 PMCID: PMC6356732 DOI: 10.3390/cells8010071] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/10/2019] [Accepted: 01/15/2019] [Indexed: 12/11/2022] Open
Abstract
Prohibitin 1 (PHB1) and prohibitin 2 (PHB2) are proteins that are ubiquitously expressed, and are present in the nucleus, cytosol, and mitochondria. Depending on the cellular localization, PHB1 and PHB2 have distinctive functions, but more evidence suggests a critical role within mitochondria. In fact, PHB proteins are highly expressed in cells that heavily depend on mitochondrial function. In mitochondria, these two proteins assemble at the inner membrane to form a supra-macromolecular structure, which works as a scaffold for proteins and lipids regulating mitochondrial metabolism, including bioenergetics, biogenesis, and dynamics in order to determine the cell fate, death, or life. PHB alterations have been found in aging and cancer, as well as neurodegenerative, cardiac, and kidney diseases, in which significant mitochondrial impairments have been observed. The molecular mechanisms by which prohibitins regulate mitochondrial function and their role in pathology are reviewed and discussed herein.
Collapse
Affiliation(s)
- Anna Signorile
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari "Aldo Moro", 70124 Bari, Italy.
| | - Giuseppe Sgaramella
- Water Research Institute (IRSA), National Research Council (CNR), Viale F. De Blasio, 5, 70132 Bari, Italy.
| | - Francesco Bellomo
- Laboratory of Nephrology, Department of Rare Diseases, Bambino Gesù Children's Hospital, Viale di S. Paolo, 15, 00149 Rome, Italy.
| | - Domenico De Rasmo
- Institute of Biomembrane, Bioenergetics and Molecular Biotechnology (IBIOM), National Research Council (CNR), 70126 Bari, Italy.
| |
Collapse
|
38
|
Hagmann H, Brinkkoetter PT. Experimental Models to Study Podocyte Biology: Stock-Taking the Toolbox of Glomerular Research. Front Pediatr 2018; 6:193. [PMID: 30057894 PMCID: PMC6053518 DOI: 10.3389/fped.2018.00193] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/11/2018] [Indexed: 01/17/2023] Open
Abstract
Diseases affecting the glomeruli of the kidney, the renal filtration units, are a leading cause of chronic kidney disease and end-stage renal failure. Despite recent advances in the understanding of glomerular biology, treatment of these disorders has remained extraordinarily challenging in many cases. The use of experimental models has proven invaluable to study renal, and in particular, glomerular biology and disease. Over the past 15 years, studies identified different and very distinct pathogenic mechanisms that result in damage, loss of glomerular visceral epithelial cells (podocytes) and progressive renal disease. However, animal studies and, in particular, mouse studies are often protracted and cumbersome due to the long reproductive cycle and high keeping costs. Transgenic and heterologous expression models have been speeded-up by novel gene editing techniques, yet they still take months. In addition, given the complex cellular biology of the filtration barrier, certain questions may not be directly addressed using mouse models due to the limited accessibility of podocytes for analysis and imaging. In this review, we will describe alternative models to study podocyte biology experimentally. We specifically discuss current podocyte cell culture models, their role in experimental strategies to analyze pathophysiologic mechanisms as well as limitations with regard to transferability of results. We introduce current models in Caenorhabditis elegans, Drosophila melanogaster, and Danio rerio that allow for analysis of protein interactions, and principle signaling pathways in functional biological structures, and enable high-throughput transgenic expression or compound screens in multicellular organisms.
Collapse
Affiliation(s)
| | - Paul T. Brinkkoetter
- Department II of Internal Medicine, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
39
|
Wischhof L, Gioran A, Sonntag-Bensch D, Piazzesi A, Stork M, Nicotera P, Bano D. A disease-associated Aifm1 variant induces severe myopathy in knockin mice. Mol Metab 2018; 13:10-23. [PMID: 29780003 PMCID: PMC6026322 DOI: 10.1016/j.molmet.2018.05.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 05/02/2018] [Accepted: 05/03/2018] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Mutations in the AIFM1 gene have been identified in recessive X-linked mitochondrial diseases. Functional and molecular consequences of these pathogenic AIFM1 mutations have been poorly studied in vivo. METHODS/RESULTS Here we provide evidence that the disease-associated apoptosis-inducing factor (AIF) deletion arginine 201 (R200 in rodents) causes pathology in knockin mice. Within a few months, posttranslational loss of the mutant AIF protein induces severe myopathy associated with a lower number of cytochrome c oxidase-positive muscle fibers. At a later stage, Aifm1 (R200 del) knockin mice manifest peripheral neuropathy, but they do not show neurodegenerative processes in the cerebellum, as observed in age-matched hypomorphic Harlequin (Hq) mutant mice. Quantitative proteomic and biochemical data highlight common molecular signatures of mitochondrial diseases, including aberrant folate-driven one-carbon metabolism and sustained Akt/mTOR signaling. CONCLUSION Our findings indicate metabolic defects and distinct tissue-specific vulnerability due to a disease-causing AIFM1 mutation, with many pathological hallmarks that resemble those seen in patients.
Collapse
Affiliation(s)
- Lena Wischhof
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Anna Gioran
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Antonia Piazzesi
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Miriam Stork
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| |
Collapse
|
40
|
Lay AC, Coward RJM. The Evolving Importance of Insulin Signaling in Podocyte Health and Disease. Front Endocrinol (Lausanne) 2018; 9:693. [PMID: 30524379 PMCID: PMC6258712 DOI: 10.3389/fendo.2018.00693] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/05/2018] [Indexed: 12/17/2022] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage renal disease worldwide, occuring in approximately one-third of diabetic patients. One of the earliest hallmarks of DKD is albuminuria, often occurring following disruptions to the glomerular filtration barrier. Podocytes are highly specialized cells with a central role in filtration barrier maintenance; hence, podocyte dysfunction is a major cause of albuminuria in many settings, including DKD. Numerous studies over the last decade have highlighted the importance of intact podocyte insulin responses in the maintenance of podocyte function. This review summarizes our current perspectives on podocyte insulin signaling, highlighting evidence to support the notion that dysregulated podocyte insulin responses contribute toward podocyte damage, particularly during the pathogenesis of DKD.
Collapse
|
41
|
BRD7 regulates the insulin-signaling pathway by increasing phosphorylation of GSK3β. Cell Mol Life Sci 2017; 75:1857-1869. [PMID: 29127434 DOI: 10.1007/s00018-017-2711-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/01/2017] [Accepted: 11/03/2017] [Indexed: 10/18/2022]
Abstract
Reduced hepatic expression levels of bromodomain-containing protein 7 (BRD7) have been suggested to play a role in the development of glucose intolerance in obesity. However, the molecular mechanism by which BRD7 regulates glucose metabolism has remained unclear. Here, we show that BRD7 increases phosphorylation of glycogen synthase kinase 3β (GSK3β) in response to activation of the insulin receptor-signaling pathway shortly after insulin stimulation and the nutrient-sensing pathway after feeding. BRD7 mediates phosphorylation of GSK3β at the Serine 9 residue and this effect on GSK3β occurs even in the absence of AKT activity. Using both in vitro and in vivo models, we further demonstrate that BRD7 mediates phosphorylation of ribosomal protein S6 kinase (S6K) and leads to increased phosphorylation of the eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) and, therefore, relieves its inhibition of the eukaryotic translation initiation factor 4E (eIF4E). However, the increase in phosphorylation of 4E-BP1 with BRD7 overexpression is blunted in the absence of AKT activity. In addition, using liver-specific BRD7 knockout (LBKO) mice, we show that BRD7 is required for mTORC1 activity on its downstream molecules. These findings show a novel basis for understanding the molecular dynamics of glucose metabolism and suggest the unique function of BRD7 in the regulation of glucose homeostasis.
Collapse
|
42
|
Chu AY, Tin A, Schlosser P, Ko YA, Qiu C, Yao C, Joehanes R, Grams ME, Liang L, Gluck CA, Liu C, Coresh J, Hwang SJ, Levy D, Boerwinkle E, Pankow JS, Yang Q, Fornage M, Fox CS, Susztak K, Köttgen A. Epigenome-wide association studies identify DNA methylation associated with kidney function. Nat Commun 2017; 8:1286. [PMID: 29097680 PMCID: PMC5668367 DOI: 10.1038/s41467-017-01297-7] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 09/05/2017] [Indexed: 11/10/2022] Open
Abstract
Chronic kidney disease (CKD) is defined by reduced estimated glomerular filtration rate (eGFR). Previous genetic studies have implicated regulatory mechanisms contributing to CKD. Here we present epigenome-wide association studies of eGFR and CKD using whole-blood DNA methylation of 2264 ARIC Study and 2595 Framingham Heart Study participants to identify epigenetic signatures of kidney function. Of 19 CpG sites significantly associated (P < 1e-07) with eGFR/CKD and replicated, five also associate with renal fibrosis in biopsies from CKD patients and show concordant DNA methylation changes in kidney cortex. Lead CpGs at PTPN6/PHB2, ANKRD11, and TNRC18 map to active enhancers in kidney cortex. At PTPN6/PHB2 cg19942083, methylation in kidney cortex associates with lower renal PTPN6 expression, higher eGFR, and less renal fibrosis. The regions containing the 243 eGFR-associated (P < 1e-05) CpGs are significantly enriched for transcription factor binding sites of EBF1, EP300, and CEBPB (P < 5e-6). Our findings highlight kidney function associated epigenetic variation. Genome-wide association studies of kidney function show enrichment of associated genetic variants in regulatory regions. Here, the authors perform epigenome-wide association studies of kidney function and disease, identifying 19 CpG sites significantly associated with these.
Collapse
Affiliation(s)
- Audrey Y Chu
- The Population Sciences Branch, Division of Intramural Research, NHLBI, NIH, Bethesda, MD, 20892, USA.,NHLBI's Framingham Heart Study, Framingham, MA, 01702, USA
| | - Adrienne Tin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Pascal Schlosser
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center-University of Freiburg, 79106, Freiburg, Germany
| | - Yi-An Ko
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Chengxiang Qiu
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Chen Yao
- The Population Sciences Branch, Division of Intramural Research, NHLBI, NIH, Bethesda, MD, 20892, USA.,NHLBI's Framingham Heart Study, Framingham, MA, 01702, USA
| | - Roby Joehanes
- The Population Sciences Branch, Division of Intramural Research, NHLBI, NIH, Bethesda, MD, 20892, USA.,NHLBI's Framingham Heart Study, Framingham, MA, 01702, USA.,Institute of Aging Research, Hebrew Senior Life, Boston, MA, 02131, USA.,Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Morgan E Grams
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Liming Liang
- Department of Biostatistics, Harvard University School of Public Health, Boston, MA, 02115, USA
| | - Caroline A Gluck
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Chunyu Liu
- The Population Sciences Branch, Division of Intramural Research, NHLBI, NIH, Bethesda, MD, 20892, USA.,NHLBI's Framingham Heart Study, Framingham, MA, 01702, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Shih-Jen Hwang
- The Population Sciences Branch, Division of Intramural Research, NHLBI, NIH, Bethesda, MD, 20892, USA.,NHLBI's Framingham Heart Study, Framingham, MA, 01702, USA
| | - Daniel Levy
- The Population Sciences Branch, Division of Intramural Research, NHLBI, NIH, Bethesda, MD, 20892, USA.,NHLBI's Framingham Heart Study, Framingham, MA, 01702, USA
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center, Houston, TX, 77030, USA
| | - James S Pankow
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, 55454, USA
| | - Qiong Yang
- NHLBI's Framingham Heart Study, Framingham, MA, 01702, USA.,Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Myriam Fornage
- Human Genetics Center, University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Caroline S Fox
- The Population Sciences Branch, Division of Intramural Research, NHLBI, NIH, Bethesda, MD, 20892, USA.,NHLBI's Framingham Heart Study, Framingham, MA, 01702, USA
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Anna Köttgen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA. .,Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center-University of Freiburg, 79106, Freiburg, Germany.
| |
Collapse
|
43
|
Lay AC, Hurcombe JA, Betin VMS, Barrington F, Rollason R, Ni L, Gillam L, Pearson GME, Østergaard MV, Hamidi H, Lennon R, Welsh GI, Coward RJM. Prolonged exposure of mouse and human podocytes to insulin induces insulin resistance through lysosomal and proteasomal degradation of the insulin receptor. Diabetologia 2017; 60:2299-2311. [PMID: 28852804 PMCID: PMC6448913 DOI: 10.1007/s00125-017-4394-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 06/28/2017] [Indexed: 12/12/2022]
Abstract
AIMS/HYPOTHESIS Podocytes are insulin-responsive cells of the glomerular filtration barrier and are key in preventing albuminuria, a hallmark feature of diabetic nephropathy. While there is evidence that a loss of insulin signalling to podocytes is detrimental, the molecular mechanisms underpinning the development of podocyte insulin resistance in diabetes remain unclear. Thus, we aimed to further investigate podocyte insulin responses early in the context of diabetic nephropathy. METHODS Conditionally immortalised human and mouse podocyte cell lines and glomeruli isolated from db/db DBA/2J mice were studied. Podocyte insulin responses were investigated with western blotting, cellular glucose uptake assays and automated fluorescent imaging of the actin cytoskeleton. Quantitative (q)RT-PCR was employed to investigate changes in mRNA. Human cell lines stably overproducing the insulin receptor (IR) and nephrin were also generated, using lentiviral constructs. RESULTS Podocytes exposed to a diabetic environment (high glucose, high insulin and the proinflammatory cytokines TNF-α and IL-6) become insulin resistant with respect to glucose uptake and activation of phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) signalling. These podocytes lose expression of the IR as a direct consequence of prolonged exposure to high insulin concentrations, which causes an increase in IR protein degradation via a proteasome-dependent and bafilomycin-sensitive pathway. Reintroducing the IR into insulin-resistant human podocytes rescues upstream phosphorylation events, but not glucose uptake. Stable expression of nephrin is also required for the insulin-stimulated glucose uptake response in podocytes and for efficient insulin-stimulated remodelling of the actin cytoskeleton. CONCLUSIONS/INTERPRETATION Together, these results suggest that IR degradation, caused by high levels of insulin, drives early podocyte insulin resistance, and that both the IR and nephrin are required for full insulin sensitivity of this cell. This could be highly relevant for the development of nephropathy in individuals with type 2 diabetes, who are commonly hyperinsulinaemic in the early phases of their disease.
Collapse
Affiliation(s)
- Abigail C Lay
- Bristol Renal, Bristol Medical School, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK
| | - Jenny A Hurcombe
- Bristol Renal, Bristol Medical School, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK
| | - Virginie M S Betin
- Bristol Renal, Bristol Medical School, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK
| | - Fern Barrington
- Bristol Renal, Bristol Medical School, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK
| | - Ruth Rollason
- Bristol Renal, Bristol Medical School, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK
| | - Lan Ni
- Bristol Renal, Bristol Medical School, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK
| | - Lawrence Gillam
- Bristol Renal, Bristol Medical School, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK
| | - Grace M E Pearson
- Bristol Renal, Bristol Medical School, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK
| | - Mette V Østergaard
- Bristol Renal, Bristol Medical School, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK
- Global Research, Novo Nordisk A/S, Måløv, Denmark
| | - Hellyeh Hamidi
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Rachel Lennon
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Gavin I Welsh
- Bristol Renal, Bristol Medical School, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK
| | - Richard J M Coward
- Bristol Renal, Bristol Medical School, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK.
| |
Collapse
|
44
|
Wang D, Zhao YQ, Han YL, Hou CC, Zhu JQ. Characterization of mitochondrial prohibitin from Boleophthalmus pectinirostris and evaluation of its possible role in spermatogenesis. FISH PHYSIOLOGY AND BIOCHEMISTRY 2017; 43:1299-1313. [PMID: 28501977 DOI: 10.1007/s10695-017-0373-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/19/2017] [Indexed: 06/07/2023]
Abstract
Prohibitin (PHB) is an evolutionarily conserved mitochondrial membrane protein. It plays a vital role in cell proteolysis, senescence, and apoptosis and is associated with spermatogenesis and sperm quality control in mammals. To study the characteristics of the PHB gene and its potential roles during spermatogenesis in Boleophthalmus pectinirostris, we cloned a 1153-bp full-length cDNA from the testis of B. pectinirostris with an open reading frame of 816 bp, which encodes 272 amino acid residues. Real-time quantitative PCR (qPCR) analysis revealed the presence of phb mRNA in all the tissues examined, with higher expression levels found in the testis, kidney, intestine, and muscle tissues. We examined the localization of phb mRNA during spermatogenesis by in situ hybridization (ISH), showing that phb mRNA was distributed in the periphery of the nucleus in primary and secondary spermatocytes. In spermatid and mature sperm, the phb mRNA gradually moved toward one side, where the flagellum is formed. Immunofluorescence (IF) results showed co-localization of the PHB and mitochondria at different stages during spermatogenesis of B. pectinirostris. The signals obtained for PHB decreased as spermatogenesis proceeded; the strongest detection signal was found in secondary spermatocytes, with lower levels of staining in other stages. Additionally, in the mature germ cells, the PHB signals were weak and aggregate in the midpiece of the flagellum.
Collapse
Affiliation(s)
- Di Wang
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China
| | - Yong-Qiang Zhao
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China
| | - Ying-Li Han
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China
| | - Cong-Cong Hou
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China.
| | - Jun-Quan Zhu
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China.
| |
Collapse
|
45
|
Koehler S, Brähler S, Braun F, Hagmann H, Rinschen MM, Späth MR, Höhne M, Wunderlich FT, Schermer B, Benzing T, Brinkkoetter PT. Construction of a viral T2A-peptide based knock-in mouse model for enhanced Cre recombinase activity and fluorescent labeling of podocytes. Kidney Int 2017; 91:1510-1517. [PMID: 28187984 DOI: 10.1016/j.kint.2016.12.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 11/23/2016] [Accepted: 12/01/2016] [Indexed: 12/16/2022]
Abstract
Podocyte injury is a key event in glomerular disease leading to proteinuria and opening the path toward glomerular scarring. As a consequence, glomerular research strives to discover molecular mechanisms and signaling pathways affecting podocyte health. The hNphs2.Cre mouse model has been a valuable tool to manipulate podocyte-specific genes and to label podocytes for lineage tracing and purification. Here we designed a novel podocyte-specific tricistronic Cre mouse model combining codon improved Cre expression and fluorescent cell labeling with mTomato under the control of the endogenous Nphs2 promoter using viral T2A-peptides. Independent expression of endogenous podocin, codon improved Cre, and mTomato was confirmed by immunofluorescence, fluorescent activated cell sorting and protein analyses. Nphs2pod.T2A.ciCre.T2A.mTomato/wild-type mice developed normally and did not show any signs of glomerular disease or off-target effects under basal conditions and in states of disease. Nphs2pod.T2A.ciCre.T2A.mTomato/wild-type-mediated gene recombination was superior to conventional hNphs2.Cre mice-mediated gene recombination. Last, we compared Cre efficiency in a disease model by mating Nphs2pod.T2A.ciCre.T2A.mTomato/wild-type and hNphs2.Cre mice to Phb2fl/fl mice. The podocyte-specific Phb2 knockout by Nphs2pod.T2A.ciCre.T2A.mTomato/wild-type mice resulted in an aggravated glomerular injury as compared to a podocyte-specific Phb2 gene deletion triggered by hNphs2.Cre. Thus, we generated the first tricistronic podocyte mouse model combining enhanced Cre recombinase efficiency and fluorescent labeling in podocytes without the need for additional matings with conventional reporter mouse lines.
Collapse
Affiliation(s)
- Sybille Koehler
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Sebastian Brähler
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Fabian Braun
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Henning Hagmann
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Markus M Rinschen
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Martin R Späth
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Martin Höhne
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany; Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | | | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany; Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany; Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | - Paul T Brinkkoetter
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
46
|
Ising C, Bharill P, Brinkkoetter S, Brähler S, Schroeter C, Koehler S, Hagmann H, Merkwirth C, Höhne M, Müller RU, Fabretti F, Schermer B, Bloch W, Kerjaschki D, Kurschat CE, Benzing T, Brinkkoetter PT. Prohibitin-2 Depletion Unravels Extra-Mitochondrial Functions at the Kidney Filtration Barrier. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 186:1128-39. [PMID: 27105734 DOI: 10.1016/j.ajpath.2015.12.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 12/16/2015] [Accepted: 12/22/2015] [Indexed: 10/21/2022]
Abstract
Mitochondrial fusion is essential for maintenance of mitochondrial function and requires the prohibitin ring complex subunit prohibitin-2 (PHB2) at the mitochondrial inner membrane. Loss of the stomatin/PHB/flotillin/HflK/C (SPFH) domain containing protein PHB2 causes mitochondrial dysfunction and defective mitochondria-mediated signaling, which is implicated in a variety of human diseases, including progressive renal disease. Here, we provide evidence of additional, extra-mitochondrial functions of this membrane-anchored protein. Immunofluorescence and immunogold labeling detected PHB2 at mitochondrial membranes and at the slit diaphragm, a specialized cell junction at the filtration slit of glomerular podocytes. PHB2 coprecipitated with podocin, another SPFH domain-containing protein, essential for the assembly of the slit diaphragm protein-lipid supercomplex. Consistent with an evolutionarily conserved extra-mitochondrial function, the ortholog of PHB2 in Caenorhabditis elegans was also not restricted to mitochondria but colocalized with the mechanosensory complex that requires the podocin ortholog MEC2 for assembly. Knockdown of phb-2 partially phenocopied loss of mec-2 in touch neurons of the nematode, resulting in impaired gentle touch sensitivity. Collectively, these data indicate that, besides its established role in mitochondria, PHB2 may have an additional function in conserved protein-lipid complexes at the plasma membrane.
Collapse
Affiliation(s)
- Christina Ising
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri
| | - Puneet Bharill
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Sibylle Brinkkoetter
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Sebastian Brähler
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Christina Schroeter
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Sybille Koehler
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Henning Hagmann
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Carsten Merkwirth
- Institute for Genetics, University of Cologne, Cologne, Germany; Howard Hughes Medical Institute, University of California Berkeley, Berkeley, California
| | - Martin Höhne
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Systems Biology of Ageing Cologne (Sybacol), Cologne, Germany
| | - Roman U Müller
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Systems Biology of Ageing Cologne (Sybacol), Cologne, Germany
| | - Francesca Fabretti
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Systems Biology of Ageing Cologne (Sybacol), Cologne, Germany
| | - Wilhelm Bloch
- Institute of Cardiovascular Research and Sport Medicine, Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Cologne, Germany
| | - Dontscho Kerjaschki
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Christine E Kurschat
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Systems Biology of Ageing Cologne (Sybacol), Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Systems Biology of Ageing Cologne (Sybacol), Cologne, Germany
| | - Paul T Brinkkoetter
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
47
|
Kuczkowski A, Brinkkoetter PT. Metabolism and homeostasis in the kidney: metabolic regulation through insulin signaling in the kidney. Cell Tissue Res 2017; 369:199-210. [DOI: 10.1007/s00441-017-2619-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 03/22/2017] [Indexed: 02/07/2023]
|
48
|
Prohibitin Signaling at the Kidney Filtration Barrier. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:563-575. [PMID: 28551807 DOI: 10.1007/978-3-319-55330-6_29] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The kidney filtration barrier consists of three well-defined anatomic layers comprising a fenestrated endothelium, the glomerular basement membrane (GBM) and glomerular epithelial cells, the podocytes. Podocytes are post-mitotic and terminally differentiated cells with primary and secondary processes. The latter are connected by a unique cell-cell contact, the slit diaphragm. Podocytes maintain the GBM and seal the kidney filtration barrier to prevent the onset of proteinuria. Loss of prohibitin-1/2 (PHB1/2) in podocytes results not only in a disturbed mitochondrial structure but also in an increased insulin/IGF-1 signaling leading to mTOR activation and a detrimental metabolic switch. As a consequence, PHB-knockout podocytes develop proteinuria and glomerulosclerosis and eventually loss of renal function. In addition, experimental evidence suggests that PHB1/2 confer additional, extra-mitochondrial functions in podocytes as they localize to the slit diaphragm and thereby stabilize the unique intercellular contact between podocytes required to maintain an effective filtration barrier.
Collapse
|
49
|
Gnudi L, Coward RJM, Long DA. Diabetic Nephropathy: Perspective on Novel Molecular Mechanisms. Trends Endocrinol Metab 2016; 27:820-830. [PMID: 27470431 DOI: 10.1016/j.tem.2016.07.002] [Citation(s) in RCA: 237] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 07/04/2016] [Accepted: 07/07/2016] [Indexed: 02/07/2023]
Abstract
Diabetes mellitus (DM) is the major cause of end-stage renal disease (ESRD) globally, and novel treatments are urgently needed. Current therapeutic approaches for diabetic nephropathy (DN) are focussing on blood pressure control with inhibitors of the renin-angiotensin-aldosterone system, on glycaemic and lipid control, and life-style changes. In this review, we highlight new molecular insights aiding our understanding of the initiation and progression of DN, including glomerular insulin resistance, dysregulation of cellular substrate utilisation, podocyte-endothelial communication, and inhibition of tubular sodium coupled glucose reabsorption. We believe that these mechanisms offer new therapeutic targets that can be exploited to develop important renoprotective treatments for DN over the next decade.
Collapse
Affiliation(s)
- Luigi Gnudi
- Cardiovascular Division, King's College London, London, SE1 9NH, UK.
| | - Richard J M Coward
- Academic Renal Unit, Dorothy Hodgkin Building, University of Bristol, Whitson Street, Bristol BS1 3NY, UK
| | - David A Long
- Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, WC1N 1EH, UK.
| |
Collapse
|
50
|
Moncunill-Massaguer C, Saura-Esteller J, Pérez-Perarnau A, Palmeri CM, Núñez-Vázquez S, Cosialls AM, González-Gironès DM, Pomares H, Korwitz A, Preciado S, Albericio F, Lavilla R, Pons G, Langer T, Iglesias-Serret D, Gil J. A novel prohibitin-binding compound induces the mitochondrial apoptotic pathway through NOXA and BIM upregulation. Oncotarget 2016; 6:41750-65. [PMID: 26497683 PMCID: PMC4747186 DOI: 10.18632/oncotarget.6154] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 09/30/2015] [Indexed: 01/08/2023] Open
Abstract
We previously described diaryl trifluorothiazoline compound 1a (hereafter referred to as fluorizoline) as a first-in-class small molecule that induces p53-independent apoptosis in a wide range of tumor cell lines. Fluorizoline directly binds to prohibitin 1 and 2 (PHBs), two proteins involved in the regulation of several cellular processes, including apoptosis. Here we demonstrate that fluorizoline-induced apoptosis is mediated by PHBs, as cells depleted of these proteins are highly resistant to fluorizoline treatment. In addition, BAX and BAK are necessary for fluorizoline-induced cytotoxic effects, thereby proving that apoptosis occurs through the intrinsic pathway. Expression analysis revealed that fluorizoline induced the upregulation of Noxa and Bim mRNA levels, which was not observed in PHB-depleted MEFs. Finally, Noxa−/−/Bim−/− MEFs and NOXA-downregulated HeLa cells were resistant to fluorizoline-induced apoptosis. All together, these findings show that fluorizoline requires PHBs to execute the mitochondrial apoptotic pathway.
Collapse
Affiliation(s)
- Cristina Moncunill-Massaguer
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - José Saura-Esteller
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - Alba Pérez-Perarnau
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - Claudia Mariela Palmeri
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - Sonia Núñez-Vázquez
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - Ana M Cosialls
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - Diana M González-Gironès
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - Helena Pomares
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - Anne Korwitz
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Sara Preciado
- Barcelona Science Park and CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, Barcelona, Spain
| | - Fernando Albericio
- Barcelona Science Park and CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, Barcelona, Spain.,Institute for Research in Biomedicine Barcelona, Barcelona, Spain.,Department of Organic Chemistry, University of Barcelona, Barcelona, Spain
| | - Rodolfo Lavilla
- Barcelona Science Park and CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, Barcelona, Spain.,Laboratory of Organic Chemistry, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Gabriel Pons
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - Thomas Langer
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Daniel Iglesias-Serret
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| | - Joan Gil
- Departament de Ciències Fisiològiques II, Universitat de Barcelona-Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Catalunya, Spain
| |
Collapse
|