1
|
Pollak MR, Friedman DJ. APOL1-associated kidney disease: modulators of the genotype-phenotype relationship. Curr Opin Nephrol Hypertens 2025; 34:191-198. [PMID: 40047214 DOI: 10.1097/mnh.0000000000001068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
PURPOSE OF REVIEW Apolipoprotein-L1 (APOL1) G1 and G2 risk variants, found in people of recent west sub-Saharan African ancestry, dramatically increase the likelihood of kidney disease, yet the incomplete penetrance an diverse clinical manifestations underscore the need to understand the molecular and environmental factors that modulate APOL1-mediated toxicity. RECENT FINDINGS Recent studies confirm that risk variants exert a toxic gain-of-function effect, exacerbated by inflammatory triggers such as HIV infection and COVID-19. Epigenetic mechanisms and microRNA pathways further modulate APOL1 expression, influencing disease penetrance. Multiple models have clarified how subcellular localization, signal peptide processing, and interactions with the endoplasmic reticulum may contribute to pathogenesis. Therapeutic advances include inhibitors targeting APOL1 ion channel activity and strategies that block key inflammatory signaling pathways. SUMMARY These findings highlight a multifaceted disease process driven by both the intrinsic toxic potential of APOL1 variants and numerous extrinsic triggers. Understanding this complex interplay will be pivotal for risk stratification and the development of precision therapies, potentially improving outcomes for populations disproportionately affected by APOL1-associated kidney disease.
Collapse
Affiliation(s)
- Martin R Pollak
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
2
|
Gęgotek A, Skrzydlewska E, Groth M, Czupryna P, Moniuszko-Malinowska A. Changes in the serum proteome profile of patients with neuroborreliosis, foresters, and patients treated according to ILADS method. Microb Pathog 2024; 197:107094. [PMID: 39486554 DOI: 10.1016/j.micpath.2024.107094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
OBJECTIVES The aim of study was to evaluate the changes in proteomic profile of human serum induced by the development of tick-borne neuroborreliosis (NB), before/after therapy, patients treated with prolonged multidrug therapy according to ILADS (International Lyme and Associated Diseases Society), and foresters frequently exposed to tick bites. METHODS A proteomics approach was used to analyze the expression of proteins in serum of patients and sex/age-matched healthy donors. The analysis was performed using SDS-PAGE/LC-MS/MS (Q-Exactive OrbiTrap mass spectrometer). RESULTS Obtained results indicated changes in the serum proteome of patients with NB putting attention to the proteins involved mainly in calcium transport/metabolism and signaling molecules that differ patients before and after classic therapy. Moreover, ILADS treated patients have different protein distribution than patients from other groups, what is the consequence of prolonged antibiotic therapy. In the case of foresters, the most important result is the increased β-secretase level. CONCLUSIONS Obtained results may contribute to a better understanding of the mechanism of the development of tick-borne diseases, as well as will allow create new opportunities for its rapid and more effective therapy. However, further studies, on larger patients groups, are needed to apply them in clinical practice.
Collapse
Affiliation(s)
- Agnieszka Gęgotek
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland.
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland.
| | - Monika Groth
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Zurawia 14, 15-540, Bialystok, Poland.
| | - Piotr Czupryna
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Zurawia 14, 15-540, Bialystok, Poland.
| | - Anna Moniuszko-Malinowska
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Zurawia 14, 15-540, Bialystok, Poland.
| |
Collapse
|
3
|
Kononikhin AS, Starodubtseva NL, Brzhozovskiy AG, Tokareva AO, Kashirina DN, Zakharova NV, Bugrova AE, Indeykina MI, Pastushkova LK, Larina IM, Mitkevich VA, Makarov AA, Nikolaev EN. Absolute Quantitative Targeted Monitoring of Potential Plasma Protein Biomarkers: A Pilot Study on Healthy Individuals. Biomedicines 2024; 12:2403. [PMID: 39457715 PMCID: PMC11504031 DOI: 10.3390/biomedicines12102403] [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: 09/25/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES The development of blood tests for the early detection of individual predisposition to socially significant diseases remains a pressing issue. METHODS In this pilot study, multiple reaction monitoring mass spectrometry (MRM-MS) with a BAK-270 assay was applied for protein concentrations analysis in blood plasma from 21 healthy volunteers of the European cohort. RESULTS The levels of 138 plasma proteins were reliably and precisely quantified in no less than 50% of samples. The quantified proteins included 66 FDA-approved markers of cardiovascular diseases (CVD), and other potential biomarkers of pathologies such as cancer, diabetes mellitus, and Alzheimer's disease. The analysis of individual variations of the plasma proteins revealed significant differences between the male (11) and female (10) groups. In total, fifteen proteins had a significantly different concentration in plasma; this included four proteins that exhibited changes greater than ±1.5-fold, three proteins (RBP4, APCS, and TTR) with higher levels in males, and one (SHBG) elevated in females. The obtained results demonstrated considerable agreement with the data collected from 20 samples of a North American cohort, which were analyzed with the similar MRM assay. The most significant differences between the cohorts of the two continents were observed in the level of 42 plasma proteins (including 24 FDA markers), of which 17 proteins showed a ≥1.5-fold change, and included proteins increased in North Americans (APOB, CRTAC1, C1QB, C1QC, C9, CRP, HP, IGHG1, IGKV4-1, SERPING1, RBP4, and AZGP1), as well as those elevated in Europeans (APOF, CD5L, HBG2, SELPLG, and TNA). CONCLUSIONS The results suggest a different contribution of specific (patho)physiological pathways (e.g., immune system and blood coagulation) to the development of socially significant diseases in Europeans and North Americans, and they should be taken into account when refining diagnostic panels.
Collapse
Affiliation(s)
- Alexey S. Kononikhin
- Project Center of Advanced Mass Spectrometry Technologies, 121205 Moscow, Russia;
- Institute of Biomedical Problems, Russian Federation State Scientific Research Center, Russian Academy of Sciences, 123007 Moscow, Russia; (D.N.K.); (L.K.P.); (I.M.L.)
| | - Natalia L. Starodubtseva
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of Russia, 117997 Moscow, Russia; (N.L.S.); (A.O.T.); (A.E.B.)
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Alexander G. Brzhozovskiy
- Project Center of Advanced Mass Spectrometry Technologies, 121205 Moscow, Russia;
- Institute of Biomedical Problems, Russian Federation State Scientific Research Center, Russian Academy of Sciences, 123007 Moscow, Russia; (D.N.K.); (L.K.P.); (I.M.L.)
| | - Alisa O. Tokareva
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of Russia, 117997 Moscow, Russia; (N.L.S.); (A.O.T.); (A.E.B.)
| | - Daria N. Kashirina
- Institute of Biomedical Problems, Russian Federation State Scientific Research Center, Russian Academy of Sciences, 123007 Moscow, Russia; (D.N.K.); (L.K.P.); (I.M.L.)
| | - Natalia V. Zakharova
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia; (N.V.Z.); (M.I.I.)
| | - Anna E. Bugrova
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of Russia, 117997 Moscow, Russia; (N.L.S.); (A.O.T.); (A.E.B.)
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia; (N.V.Z.); (M.I.I.)
| | - Maria I. Indeykina
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia; (N.V.Z.); (M.I.I.)
| | - Liudmila Kh. Pastushkova
- Institute of Biomedical Problems, Russian Federation State Scientific Research Center, Russian Academy of Sciences, 123007 Moscow, Russia; (D.N.K.); (L.K.P.); (I.M.L.)
| | - Irina M. Larina
- Institute of Biomedical Problems, Russian Federation State Scientific Research Center, Russian Academy of Sciences, 123007 Moscow, Russia; (D.N.K.); (L.K.P.); (I.M.L.)
| | - Vladimir A. Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Science, 119991 Moscow, Russia; (V.A.M.); (A.A.M.)
| | - Alexander A. Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Science, 119991 Moscow, Russia; (V.A.M.); (A.A.M.)
| | - Evgeny N. Nikolaev
- Project Center of Advanced Mass Spectrometry Technologies, 121205 Moscow, Russia;
| |
Collapse
|
4
|
Tian J, Gao J, Cheng C, Xu Z, Chen X, Wu Y, Fu G, Jin B. NOP2-mediated 5-methylcytosine modification of APOL1 messenger RNA activates PI3K-Akt and facilitates clear cell renal cell carcinoma progression. Int J Biol Sci 2024; 20:4853-4871. [PMID: 39309431 PMCID: PMC11414376 DOI: 10.7150/ijbs.97503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
Background: By regulating the functions of multiple RNAs, 5-methylcytosine (m5C) RNA methylation, particularly mediated by NOP2, is involved in tumorigenesis and developments. However, the specific functions and potential mechanisms of m5C, especially involving NOP2, in clear-cell renal cell carcinoma (ccRCC), remain unclear. Methods: NOP2 expression in cell lines and patient tissues was detected using western blotting, quantitative real-time polymerase chain reaction (RT-qPCR), and immunohistochemistry. The biological effects of NOP2 on ccRCC cells were investigated through a series of in vitro and in vivo experiments. To explore the potential regulatory mechanisms by which NOP2 affects ccRCC progression, m5C bisulfite sequencing, RNA-sequencing, RNA immunoprecipitation and methylated RNA immunoprecipitation (RIP/MeRIP) RT-qPCR assay, luciferase reporter assay, RNA stability assay, and bioinformatic analysis were performed. Results: NOP2 expression was significantly upregulated in ccRCC tissues and was associated with poor prognosis. Moreover, loss-of-function and gain-of-function assays demonstrated that NOP2 altered ccRCC cell proliferation, migration, and invasion. Mechanistically, NOP2 stimulated m5C modification of apolipoprotein L1 (APOL1) mRNA, and m5C reader YBX1 stabilized APOL1 mRNA through recognizing and binding to m5C site in the 3'-untranslated regions. Silencing APOL1 expression inhibited ccRCC cell proliferation in vitro and tumor formation in vivo. Furthermore, NOP2/APOL1 affected ccRCC progression via the PI3K-Akt signaling pathway. Conclusion: NOP2 functions as an oncogene in ccRCC by promoting tumor progression through the m5C-dependent stabilization of APOL1, which in turn regulates the PI3K-Akt signaling pathway, suggesting a potential therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Junjie Tian
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jianguo Gao
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310003, China
| | - Cheng Cheng
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310003, China
| | - Zhijie Xu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310003, China
| | - Xiaoyi Chen
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310003, China
| | - Yunfei Wu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310003, China
| | - Guanghou Fu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310003, China
| | - Baiye Jin
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310003, China
| |
Collapse
|
5
|
Langbøl M, Saruhanian A, Saruhanian S, Tiedemann D, Baskaran T, Vohra R, Rives AS, Moreira J, Prokosch V, Liu H, Lackmann JW, Müller S, Nielsen CH, Kolko M, Rovelt J. Proteomic and Cytokine Profiling in Plasma from Patients with Normal-Tension Glaucoma and Ocular Hypertension. Cell Mol Neurobiol 2024; 44:59. [PMID: 39150567 PMCID: PMC11329415 DOI: 10.1007/s10571-024-01492-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/06/2024] [Indexed: 08/17/2024]
Abstract
Primary open-angle glaucoma (POAG) is subdivided depending on eye pressure. Patients with normal-tension glaucoma (NTG) have never had high intraocular pressure (IOP) measured while patients with ocular hypertension (OHT) have high eye pressure but no signs of glaucoma. Although IOP is considered to be a risk factor for all glaucoma patients, it is reasonable to assume that other risk factors such as inflammation play a role. We aimed to characterize the proteome and cytokine profile during hypoxia in plasma from patients with NTG (n = 10), OHT (n = 10), and controls (n = 10). Participants were exposed to hypoxia for two hours, followed by 30 min of normoxia. Samples were taken before ("baseline"), during ("hypoxia"), and after hypoxia ("recovery"). Proteomics based on liquid chromatography coupled with mass spectrometry (LC-MS) was performed. Cytokines were measured by Luminex assays. Bioinformatic analyses indicated the involvement of complement and coagulation cascades in NTG and OHT. Regulation of high-density lipoprotein 3 (HDL3) apolipoproteins suggested that changes in cholesterol metabolism are related to OHT. Hypoxia decreased the level of tumor necrosis factor-α (TNF-α) in OHT patients compared to controls. Circulating levels of interleukin-1β (IL-1β) and C-reactive protein (CRP) were decreased in NTG patients compared to controls during hypoxia. After recovery, plasma interleukin-6 (IL-6) was upregulated in patients with NTG and OHT. Current results indicate an enhanced systemic immune response in patients with NTG and OHT, which correlates with pathogenic events in glaucoma. Apolipoproteins may have anti-inflammatory effects, enabling OHT patients to withstand inflammation and development of glaucoma despite high IOP.
Collapse
Affiliation(s)
- Mia Langbøl
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, Building 22, 2100, Copenhagen Ø, Denmark.
| | - Arevak Saruhanian
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, Building 22, 2100, Copenhagen Ø, Denmark
| | - Sarkis Saruhanian
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, Building 22, 2100, Copenhagen Ø, Denmark
- Department of Veterinary & Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Daniel Tiedemann
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, Building 22, 2100, Copenhagen Ø, Denmark
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet-Glostrup, Glostrup, Denmark
| | - Thisayini Baskaran
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, Building 22, 2100, Copenhagen Ø, Denmark
| | - Rupali Vohra
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, Building 22, 2100, Copenhagen Ø, Denmark
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet-Glostrup, Glostrup, Denmark
| | - Amalie Santaolalla Rives
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, Building 22, 2100, Copenhagen Ø, Denmark
| | - José Moreira
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, Building 22, 2100, Copenhagen Ø, Denmark
| | - Verena Prokosch
- Department of Ophthalmology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50937, Cologne, Germany
| | - Hanhan Liu
- Department of Ophthalmology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50937, Cologne, Germany
| | - Jan-Wilm Lackmann
- CECAD/CMMC Proteomics Facility, CECAD Research Center, University of Cologne, Cologne, Germany
| | - Stefan Müller
- CECAD/CMMC Proteomics Facility, CECAD Research Center, University of Cologne, Cologne, Germany
| | - Claus Henrik Nielsen
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, 2200, Copenhagen, Denmark
- Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, Building 22, 2100, Copenhagen Ø, Denmark
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet-Glostrup, Glostrup, Denmark
| | - Jens Rovelt
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, Building 22, 2100, Copenhagen Ø, Denmark
| |
Collapse
|
6
|
Paz-Barba M, Muñoz Garcia A, de Winter TJJ, de Graaf N, van Agen M, van der Sar E, Lambregtse F, Daleman L, van der Slik A, Zaldumbide A, de Koning EJP, Carlotti F. Apolipoprotein L genes are novel mediators of inflammation in beta cells. Diabetologia 2024; 67:124-136. [PMID: 37924378 PMCID: PMC10709252 DOI: 10.1007/s00125-023-06033-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/22/2023] [Indexed: 11/06/2023]
Abstract
AIMS/HYPOTHESIS Inflammation induces beta cell dysfunction and demise but underlying molecular mechanisms remain unclear. The apolipoprotein L (APOL) family of genes has been associated with innate immunity and apoptosis in non-pancreatic cell types, but also with metabolic syndrome and type 2 diabetes mellitus. Here, we hypothesised that APOL genes play a role in inflammation-induced beta cell damage. METHODS We used single-cell transcriptomics datasets of primary human pancreatic islet cells to study the expression of APOL genes upon specific stress conditions. Validation of the findings was carried out in EndoC-βH1 cells and primary human islets. Finally, we performed loss- and gain-of-function experiments to investigate the role of APOL genes in beta cells. RESULTS APOL genes are expressed in primary human beta cells and APOL1, 2 and 6 are strongly upregulated upon inflammation via the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway. APOL1 overexpression increases endoplasmic reticulum stress while APOL1 knockdown prevents cytokine-induced beta cell death and interferon-associated response. Furthermore, we found that APOL genes are upregulated in beta cells from donors with type 2 diabetes compared with donors without diabetes mellitus. CONCLUSIONS/INTERPRETATION APOLs are novel regulators of islet inflammation and may contribute to beta cell damage during the development of diabetes. DATA AVAILABILITY scRNAseq data generated by our laboratory and used in this study are available in the Gene Expression Omnibus (GEO; www.ncbi.nlm.nih.gov/geo/ ), accession number GSE218316.
Collapse
Affiliation(s)
- Miriam Paz-Barba
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Amadeo Muñoz Garcia
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Twan J J de Winter
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Natascha de Graaf
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Maarten van Agen
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Elisa van der Sar
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Ferdy Lambregtse
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Lizanne Daleman
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Arno van der Slik
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Arnaud Zaldumbide
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Eelco J P de Koning
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Françoise Carlotti
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
7
|
Shen C, Han C, Li Z, Yan Y, Li C, Chen H, Fan Z, Hu H. Construction and Validation of a Prognostic Model Based on Pyroptosis-related Genes in Bladder Cancer. Comb Chem High Throughput Screen 2024; 27:2335-2349. [PMID: 37849225 DOI: 10.2174/0113862073256363230929200157] [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: 04/30/2023] [Revised: 08/02/2023] [Accepted: 08/23/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Bladder cancer (BCa) is a highly prevalent disease with a poor prognosis. There is no better forecasting method for it yet. Current studies demonstrate that pyroptosis is involved in the development and progression of various cancers. METHODS This study employed bioinformatics techniques to analyze the data of BCa patients obtained from the TCGA and GEO databases in order to construct a prognostic risk model. The TCGA dataset was used for the training set, and the multiple external datasets (including GSE13507, GSE31684, GSE48075, IMvigor210, and GSE32894) were applied as the validation sets. Prognostic-associated pyroptosis genes screened by univariate Cox regression analysis were utilized to construct the lasso Cox regression model. GO and KEGG analysis results identified the selected genes that are primarily involved in the inflammation and cell death processes. The related patients were grouped into low- and high-risk groups. Kaplan-Meier survival analysis was performed to compare survival differences between the risk groups. The accuracy of this risk prediction model was assessed by ROC. We also applied the Human Protein Atlas (HPA) to detect the protein expression of these genes. Subsequently, qRT-PCR was performed to verify the expression of these model genes. RESULTS There are 29 pyroptosis-related genes with significant expression differences between BCa and corresponding adjacent tissues, and 11 genes (SH2D2A, CHMP4C, MRFAP1L1, GBP2, EHBP1, RAD9A, ANXA1, TMEM109, HEYL, APOL2, ORMDL1) were picked by univariate and LASSO Cox regression analysis. Immunological cell infiltration and ssGSEA results further indicated that the low and high-risk groups were substantially correlated with the immune status of BCa patients. According to TCGA and multiple external datasets, Kaplan-Meier survival curves showed the overall survival rate of the high-risk group to be decreased. ROC curves showed the model established to be accurate and reliable. Moreover, the HPA database also demonstrated the verification of the modeled genes' expression in BCa and normal bladder tissue using the HPA database. qRT-PCR results also suggested the up-regulated EHBP1 and down-regulated RAD9A mRNA expression levels to be confirmed in 15 pairs of BCa and corresponding adjacent tissues. CONCLUSION This study presents the development and validation of a novel gene signature associated with pyroptosis, which holds the potential for predicting patient outcomes in BCa and providing insights into the immune microenvironment of BCa.
Collapse
Affiliation(s)
- Chong Shen
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, P.R. China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, P.R. China
| | - Chenyang Han
- Department of Pain Therapeutic Centre, The Second Hospital of Tianjin Medical University, Tianjin, P.R. China
| | - Zhi Li
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, P.R. China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, P.R. China
| | - Yan Yan
- Department of Vascular Surgery, University Hospital Aachen, Pauwelsstr 30, Aachen 52074, Germany
| | - Chenyun Li
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, P.R. China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, P.R. China
| | - Houyuan Chen
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, P.R. China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, P.R. China
| | - Zhenqian Fan
- Department of Endocrinology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Hailong Hu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, P.R. China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, P.R. China
| |
Collapse
|
8
|
Lee HJ, Zhao Y, Fleming I, Mehta S, Wang X, Wyk BV, Ronca SE, Kang H, Chou CH, Fatou B, Smolen KK, Levy O, Clish CB, Xavier RJ, Steen H, Hafler DA, Love JC, Shalek AK, Guan L, Murray KO, Kleinstein SH, Montgomery RR. Early cellular and molecular signatures correlate with severity of West Nile virus infection. iScience 2023; 26:108387. [PMID: 38047068 PMCID: PMC10692672 DOI: 10.1016/j.isci.2023.108387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/04/2023] [Accepted: 10/27/2023] [Indexed: 12/05/2023] Open
Abstract
Infection with West Nile virus (WNV) drives a wide range of responses, from asymptomatic to flu-like symptoms/fever or severe cases of encephalitis and death. To identify cellular and molecular signatures distinguishing WNV severity, we employed systems profiling of peripheral blood from asymptomatic and severely ill individuals infected with WNV. We interrogated immune responses longitudinally from acute infection through convalescence employing single-cell protein and transcriptional profiling complemented with matched serum proteomics and metabolomics as well as multi-omics analysis. At the acute time point, we detected both elevation of pro-inflammatory markers in innate immune cell types and reduction of regulatory T cell activity in participants with severe infection, whereas asymptomatic donors had higher expression of genes associated with anti-inflammatory CD16+ monocytes. Therefore, we demonstrated the potential of systems immunology using multiple cell-type and cell-state-specific analyses to identify correlates of infection severity and host cellular activity contributing to an effective anti-viral response.
Collapse
Affiliation(s)
- Ho-Joon Lee
- Department of Genetics and Yale Center for Genome Analysis, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yujiao Zhao
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| | - Ira Fleming
- The Institute of Medical Science and Engineering, Department of Chemistry, and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Sameet Mehta
- Department of Genetics and Yale Center for Genome Analysis, Yale School of Medicine, New Haven, CT 06520, USA
| | - Xiaomei Wang
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| | - Brent Vander Wyk
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| | - Shannon E. Ronca
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX 77030, USA
| | - Heather Kang
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Chih-Hung Chou
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Benoit Fatou
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Kinga K. Smolen
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ofer Levy
- Department of Infectious Disease, Precision Vaccines Program, Boston Children’s Hospital, and Harvard Medical School, Boston, MA 02115, USA
| | - Clary B. Clish
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ramnik J. Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Center for Computational and Integrative Biology and Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Hanno Steen
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX 77030, USA
| | - David A. Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - J. Christopher Love
- The Institute of Medical Science and Engineering, Department of Chemistry, and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Alex K. Shalek
- The Institute of Medical Science and Engineering, Department of Chemistry, and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Leying Guan
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06520, USA
| | - Kristy O. Murray
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX 77030, USA
| | - Steven H. Kleinstein
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA
| | - Ruth R. Montgomery
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
9
|
Vasquez-Rios G, De Cos M, Campbell KN. Novel Therapies in APOL1-Mediated Kidney Disease: From Molecular Pathways to Therapeutic Options. Kidney Int Rep 2023; 8:2226-2234. [PMID: 38025220 PMCID: PMC10658239 DOI: 10.1016/j.ekir.2023.08.028] [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: 08/05/2023] [Accepted: 08/21/2023] [Indexed: 12/01/2023] Open
Abstract
Apolipoprotein L1 (APOL1) high-risk variants confer an increased risk for the development and progression of kidney disease among individuals of recent African ancestry. Over the past several years, significant progress has been made in understanding the pathogenesis of APOL1-mediated kidney diseases (AMKD), including genetic regulation, environmental interactions, immunomodulatory, proinflammatory and apoptotic signaling processes, as well as the complex role of APOL1 as an ion channel. Collectively, these findings have paved the way for novel therapeutic strategies to mitigate APOL1-mediated kidney injury. Precision medicine approaches are being developed to identify subgroups of AMKD patients who may benefit from these targeted interventions, fueling hope for improved clinical outcomes. This review summarizes key mechanistic insights in the pathogenesis of AMKD, emergent therapies, and discusses future challenges.
Collapse
Affiliation(s)
- George Vasquez-Rios
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Marina De Cos
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kirk N. Campbell
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
10
|
Rashmi P, Sigdel TK, Rychkov D, Damm I, Da Silva AA, Vincenti F, Lourenco AL, Craik CS, Reiser J, Sarwal MM. Perturbations in podocyte transcriptome and biological pathways induced by FSGS associated circulating factors. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:315. [PMID: 37404982 PMCID: PMC10316099 DOI: 10.21037/atm-22-3670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 12/03/2022] [Indexed: 07/06/2023]
Abstract
Background Focal segmental glomerulosclerosis (FSGS) is frequently associated with heavy proteinuria and progressive renal failure requiring dialysis or kidney transplantation. However, primary FSGS also has a ~40% risk of recurrence of disease in the transplanted kidney (rFSGS). Multiple circulating factors have been identified to contribute to the pathogenesis of primary and rFSGS including soluble urokinase-type plasminogen activator receptor (suPAR) and patient-derived CD40 autoantibody (CD40autoAb). However, the downstream effector pathways specific to individual factors require further study. The tumor necrosis factor, TNF pathway activation by one or more circulating factors present in the sera of patients with FSGS has been supported by multiple studies. Methods A human in vitro model was used to study podocyte injury measured as the loss of actin stress fibers. Anti-CD40 autoantibody was isolated from FSGS patients (recurrent and non-recurrent) and control patients with ESRD due to non-FSGS related causes. Two novel human antibodies-anti-uPAR (2G10) and anti-CD40 antibody (Bristol Meyer Squibb, 986090) were tested for their ability to rescue podocyte injury. Podocytes treated with patient derived antibody were transcriptionally profiled using whole human genome microarray. Results Here we show that podocyte injury caused by sera from FSGS patients is mediated by CD40 and suPAR and can be blocked by human anti-uPAR and anti-CD40 antibodies. Transcriptomic studies to compare the molecules and pathways activated in response to CD40 autoantibody from rFSGS patients (rFSGS/CD40autoAb) and suPAR, identified unique inflammatory pathways associated with FSGS injury. Conclusions We identified several novel and previously described genes associated with FSGS progression. Targeted blockade of suPAR and CD40 pathways with novel human antibodies showed inhibition of podocyte injury in FSGS.
Collapse
Affiliation(s)
- Priyanka Rashmi
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Tara K. Sigdel
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Dmitry Rychkov
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Izabella Damm
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Andrea Alice Da Silva
- Department of Immunology, Laboratory of Autoimmunity and Immunoregulation, Fluminense Federal University, Niteroi, Brazil
| | - Flavio Vincenti
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Andre L. Lourenco
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Charles S. Craik
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Minnie M. Sarwal
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
11
|
Adeva-Andany MM, Funcasta-Calderón R, Fernández-Fernández C, Ameneiros-Rodríguez E, Vila-Altesor M, Castro-Quintela E. The metabolic effects of APOL1 in humans. Pflugers Arch 2023:10.1007/s00424-023-02821-z. [PMID: 37261508 PMCID: PMC10233197 DOI: 10.1007/s00424-023-02821-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/04/2023] [Accepted: 05/21/2023] [Indexed: 06/02/2023]
Abstract
Harboring apolipoprotein L1 (APOL1) variants coded by the G1 or G2 alleles of the APOL1 gene increases the risk for collapsing glomerulopathy, focal segmental glomerulosclerosis, albuminuria, chronic kidney disease, and accelerated kidney function decline towards end-stage kidney disease. However, most subjects carrying APOL1 variants do not develop the kidney phenotype unless a second clinical condition adds to the genotype, indicating that modifying factors modulate the genotype-phenotype correlation. Subjects with an APOL1 high-risk genotype are more likely to develop essential hypertension or obesity, suggesting that carriers of APOL1 risk variants experience more pronounced insulin resistance compared to noncarriers. Likewise, arterionephrosclerosis (the pathological correlate of hypertension-associated nephropathy) and glomerulomegaly take place among carriers of APOL1 risk variants, and these pathological changes are also present in conditions associated with insulin resistance, such as essential hypertension, aging, and diabetes. Insulin resistance may contribute to the clinical features associated with the APOL1 high-risk genotype. Unlike carriers of wild-type APOL1, bearers of APOL1 variants show impaired formation of lipid droplets, which may contribute to inducing insulin resistance. Nascent lipid droplets normally detach from the endoplasmic reticulum into the cytoplasm, although the proteins that enable this process remain to be fully defined. Wild-type APOL1 is located in the lipid droplet, whereas mutated APOL1 remains sited at the endoplasmic reticulum, suggesting that normal APOL1 may participate in lipid droplet biogenesis. The defective formation of lipid droplets is associated with insulin resistance, which in turn may modulate the clinical phenotype present in carriers of APOL1 risk variants.
Collapse
Affiliation(s)
- María M Adeva-Andany
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain.
| | - Raquel Funcasta-Calderón
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain
| | - Carlos Fernández-Fernández
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain
| | - Eva Ameneiros-Rodríguez
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain
| | - Matilde Vila-Altesor
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain
| | - Elvira Castro-Quintela
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain
| |
Collapse
|
12
|
Pavanello C, Ossoli A. HDL and chronic kidney disease. ATHEROSCLEROSIS PLUS 2023; 52:9-17. [PMID: 37193017 PMCID: PMC10182177 DOI: 10.1016/j.athplu.2023.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 03/22/2023] [Accepted: 04/06/2023] [Indexed: 05/18/2023]
Abstract
Low HDL-cholesterol (HDL-C) concentrations are a typical trait of the dyslipidemia associated with chronic kidney disease (CKD). In this condition, plasma HDLs are characterized by alterations in structure and function, and these particles can lose their atheroprotective functions, e.g., the ability to promote cholesterol efflux from peripheral cells, anti-oxidant and anti-inflammatory proprieties and they can even become dysfunctional, i.e., exactly damaging. The reduction in plasma HDL-C levels appears to be the only lipid alteration clearly linked to the progression of renal disease in CKD patients. The association between the HDL system and CKD development and progression is also supported by the presence of genetic kidney alterations linked to HDL metabolism, including mutations in the APOA1, APOE, APOL and LCAT genes. Among these, renal disease associated with LCAT deficiency is well characterized and lipid abnormalities detected in LCAT deficiency carriers mirror the ones observed in CKD patients, being present also in acquired LCAT deficiency. This review summarizes the major alterations in HDL structure and function in CKD and how genetic alterations in HDL metabolism can be linked to kidney dysfunction. Finally, the possibility of targeting the HDL system as possible strategy to slow CKD progression is reviewed.
Collapse
Affiliation(s)
| | - Alice Ossoli
- Corresponding author. Center E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti”, Università degli Studi di Milano, Via G. Balzaretti, 9, 20133, Milano, Italy.
| |
Collapse
|
13
|
D’Arca D, Severi L, Ferrari S, Dozza L, Marverti G, Magni F, Chinello C, Pagani L, Tagliazucchi L, Villani M, d’Addese G, Piga I, Conteduca V, Rossi L, Gurioli G, De Giorgi U, Losi L, Costi MP. Serum Mass Spectrometry Proteomics and Protein Set Identification in Response to FOLFOX-4 in Drug-Resistant Ovarian Carcinoma. Cancers (Basel) 2023; 15:cancers15020412. [PMID: 36672361 PMCID: PMC9856519 DOI: 10.3390/cancers15020412] [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: 12/07/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Ovarian cancer is a highly lethal gynecological malignancy. Drug resistance rapidly occurs, and different therapeutic approaches are needed. So far, no biomarkers have been discovered to predict early response to therapies in the case of multi-treated ovarian cancer patients. The aim of our investigation was to identify a protein panel and the molecular pathways involved in chemotherapy response through a combination of studying proteomics and network enrichment analysis by considering a subset of samples from a clinical setting. Differential mass spectrometry studies were performed on 14 serum samples from patients with heavily pretreated platinum-resistant ovarian cancer who received the FOLFOX-4 regimen as a salvage therapy. The serum was analyzed at baseline time (T0) before FOLFOX-4 treatment, and before the second cycle of treatment (T1), with the aim of understanding if it was possible, after a first treatment cycle, to detect significant proteome changes that could be associated with patients responses to therapy. A total of 291 shared expressed proteins was identified and 12 proteins were finally selected between patients who attained partial response or no-response to chemotherapy when both response to therapy and time dependence (T0, T1) were considered in the statistical analysis. The protein panel included APOL1, GSN, GFI1, LCATL, MNA, LYVE1, ROR1, SHBG, SOD3, TEC, VPS18, and ZNF573. Using a bioinformatics network enrichment approach and metanalysis study, relationships between serum and cellular proteins were identified. An analysis of protein networks was conducted and identified at least three biological processes with functional and therapeutic significance in ovarian cancer, including lipoproteins metabolic process, structural component modulation in relation to cellular apoptosis and autophagy, and cellular oxidative stress response. Five proteins were almost independent from the network (LYVE1, ROR1, TEC, GFI1, and ZNF573). All proteins were associated with response to drug-resistant ovarian cancer resistant and were mechanistically connected to the pathways associated with cancer arrest. These results can be the basis for extending a biomarker discovery process to a clinical trial, as an early predictive tool of chemo-response to FOLFOX-4 of heavily treated ovarian cancer patients and for supporting the oncologist to continue or to interrupt the therapy.
Collapse
Affiliation(s)
- Domenico D’Arca
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125 Modena, Italy
| | - Leda Severi
- Department Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Stefania Ferrari
- Department Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Luca Dozza
- Seràgnoli Institute of Hematology, Department of Experimental, Diagnostic and Specialty Medicine, Bologna University School of Medicine, S. Orsola Malpighi Hospital, 40138 Bologna, Italy
| | - Gaetano Marverti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125 Modena, Italy
| | - Fulvio Magni
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano-Bicocca, 20126 Vedano al Lambro, Italy
| | - Clizia Chinello
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano-Bicocca, 20126 Vedano al Lambro, Italy
| | - Lisa Pagani
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano-Bicocca, 20126 Vedano al Lambro, Italy
| | - Lorenzo Tagliazucchi
- Department Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
- Clinical and Experimental Medicine (CEM) Doctorate School, University of Modena and Reggio Emilia, Via Campi 287, 41125 Modena, Italy
| | - Marco Villani
- Department of Physics, Informatics and Mathematics, Modena and Reggio Emilia University, Via Campi 213/A, 41125 Modena, Italy
| | - Gianluca d’Addese
- Department of Physics, Informatics and Mathematics, Modena and Reggio Emilia University, Via Campi 213/A, 41125 Modena, Italy
| | - Isabella Piga
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano-Bicocca, 20126 Vedano al Lambro, Italy
| | - Vincenza Conteduca
- IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), 47014 Meldola, Italy
| | - Lorena Rossi
- IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), 47014 Meldola, Italy
| | - Giorgia Gurioli
- IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), 47014 Meldola, Italy
| | - Ugo De Giorgi
- IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), 47014 Meldola, Italy
| | - Lorena Losi
- Department Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
- Correspondence: (L.L.); (M.P.C.)
| | - Maria Paola Costi
- Department Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
- Correspondence: (L.L.); (M.P.C.)
| |
Collapse
|
14
|
Wong SK, Ramli FF, Ali A, Ibrahim N‘I. Genetics of Cholesterol-Related Genes in Metabolic Syndrome: A Review of Current Evidence. Biomedicines 2022; 10:biomedicines10123239. [PMID: 36551995 PMCID: PMC9775320 DOI: 10.3390/biomedicines10123239] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Metabolic syndrome (MetS) refers to a cluster of metabolic dysregulations, which include insulin resistance, obesity, atherogenic dyslipidemia and hypertension. The complex pathogenesis of MetS encompasses the interplay between environmental and genetic factors. Environmental factors such as excessive nutrients and sedentary lifestyle are modifiable and could be improved by lifestyle modification. However, genetic susceptibility to MetS, a non-modifiable factor, has attracted the attention of researchers, which could act as the basis for future diagnosis, prognosis, and therapy for MetS. Several cholesterol-related genes associated with each characteristic of MetS have been identified, such as apolipoprotein, lipoprotein lipase (LPL), cholesteryl ester transfer protein (CETP) and adiponectin. This review aims to summarize the genetic information of cholesterol-related genes in MetS, which may potentially serve as biomarkers for early prevention and management of MetS.
Collapse
Affiliation(s)
- Sok Kuan Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia
| | - Fitri Fareez Ramli
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia
- Clinical Psychopharmacology Research Unit, Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK
| | - Adli Ali
- Department of Pediatrics, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia
| | - Nurul ‘Izzah Ibrahim
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia
- Correspondence: ; Tel.: +60-39145-9545
| |
Collapse
|
15
|
Boada P, Fatou B, Belperron AA, Sigdel TK, Smolen KK, Wurie Z, Levy O, Ronca SE, Murray KO, Liberto JM, Rashmi P, Kerwin M, Montgomery RR, Bockenstedt LK, Steen H, Sarwal MM. Longitudinal serum proteomics analyses identify unique and overlapping host response pathways in Lyme disease and West Nile virus infection. Front Immunol 2022; 13:1012824. [PMID: 36569838 PMCID: PMC9784464 DOI: 10.3389/fimmu.2022.1012824] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 11/07/2022] [Indexed: 12/14/2022] Open
Abstract
Advancement in proteomics methods for interrogating biological samples has helped identify disease biomarkers for early diagnostics and unravel underlying molecular mechanisms of disease. Herein, we examined the serum proteomes of 23 study participants presenting with one of two common arthropod-borne infections: Lyme disease (LD), an extracellular bacterial infection or West Nile virus infection (WNV), an intracellular viral infection. The LC/MS based serum proteomes of samples collected at the time of diagnosis and during convalescence were assessed using a depletion-based high-throughput shotgun proteomics (dHSP) pipeline as well as a non-depleting blotting-based low-throughput platform (MStern). The LC/MS integrated analyses identified host proteome responses in the acute and recovery phases shared by LD and WNV infections, as well as differentially abundant proteins that were unique to each infection. Notably, we also detected proteins that distinguished localized from disseminated LD and asymptomatic from symptomatic WNV infection. The proteins detected in both diseases with the dHSP pipeline identified unique and overlapping proteins detected with the non-depleting MStern platform, supporting the utility of both detection methods. Machine learning confirmed the use of the serum proteome to distinguish the infection from healthy control sera but could not develop discriminatory models between LD and WNV at current sample numbers. Our study is the first to compare the serum proteomes in two arthropod-borne infections and highlights the similarities in host responses even though the pathogens and the vectors themselves are different.
Collapse
Affiliation(s)
- Patrick Boada
- Division of Transplant Surgery, Department of Surgery, University of California, San Francisco, CA, United States
| | - Benoit Fatou
- Department of Pathology, Boston Children’s Hospital - Harvard Medical School, Boston, MA, United States
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
| | - Alexia A. Belperron
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Tara K. Sigdel
- Division of Transplant Surgery, Department of Surgery, University of California, San Francisco, CA, United States
| | - Kinga K. Smolen
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Infectious Diseases, Boston Children’s Hospital – Harvard Medical School, Boston, MA, United States
| | - Zainab Wurie
- Department of Pathology, Boston Children’s Hospital - Harvard Medical School, Boston, MA, United States
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
| | - Ofer Levy
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Infectious Diseases, Boston Children’s Hospital – Harvard Medical School, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology & Harvard, Cambridge, MA, United States
| | - Shannon E. Ronca
- Division of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
- William T. Shearer Center for Human Immunobiology, Texas Children’s Hospital, Houston, TX, United States
| | - Kristy O. Murray
- Division of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
- William T. Shearer Center for Human Immunobiology, Texas Children’s Hospital, Houston, TX, United States
| | - Juliane M. Liberto
- Division of Transplant Surgery, Department of Surgery, University of California, San Francisco, CA, United States
| | - Priyanka Rashmi
- Division of Transplant Surgery, Department of Surgery, University of California, San Francisco, CA, United States
| | - Maggie Kerwin
- Division of Transplant Surgery, Department of Surgery, University of California, San Francisco, CA, United States
| | - Ruth R. Montgomery
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Linda K. Bockenstedt
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Hanno Steen
- Department of Pathology, Boston Children’s Hospital - Harvard Medical School, Boston, MA, United States
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
| | - Minnie M. Sarwal
- Division of Transplant Surgery, Department of Surgery, University of California, San Francisco, CA, United States
| |
Collapse
|
16
|
Blazer A, Qian Y, Schlegel MP, Algasas H, Buyon JP, Cadwell K, Cammer M, Heffron SP, Liang FX, Mehta-Lee S, Niewold T, Rasmussen SE, Clancy RM. APOL1 variant-expressing endothelial cells exhibit autophagic dysfunction and mitochondrial stress. Front Genet 2022; 13:769936. [PMID: 36238153 PMCID: PMC9551299 DOI: 10.3389/fgene.2022.769936] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 08/16/2022] [Indexed: 12/09/2022] Open
Abstract
Polymorphisms in the Apolipoprotein L1 (APOL1) gene are common in ancestrally African populations, and associate with kidney injury and cardiovascular disease. These risk variants (RV) provide an advantage in resisting Trypanosoma brucei, the causal agent of African trypanosomiasis, and are largely absent from non-African genomes. Clinical associations between the APOL1 high risk genotype (HRG) and disease are stronger in those with comorbid infectious or immune disease. To understand the interaction between cytokine exposure and APOL1 cytotoxicity, we established human umbilical vein endothelial cell (HUVEC) cultures representing each APOL1 genotype. Untreated HUVECs were compared to IFNɣ-exposed; and APOL1 expression, mitochondrial function, lysosome integrity, and autophagic flux were measured. IFNɣ increased median APOL1 expression across all genotypes 22.1 (8.3 to 29.8) fold (p=0.02). Compared to zero risk variant-carrying HUVECs (0RV), HUVECs carrying 2 risk variant copies (2RV) showed both depressed baseline and maximum mitochondrial oxygen consumption (p<0.01), and impaired mitochondrial networking on MitoTracker assays. These cells also demonstrated a contracted lysosomal compartment, and an accumulation of autophagosomes suggesting a defect in autophagic flux. Upon blocking autophagy with non-selective lysosome inhibitor, hydroxychloroquine, autophagosome accumulation between 0RV HUVECs and untreated 2RV HUVECs was similar, implicating lysosomal dysfunction in the HRG-associated autophagy defect. Compared to 0RV and 2RV HUVECs, HUVECs carrying 1 risk variant copy (1RV) demonstrated intermediate mitochondrial respiration and autophagic flux phenotypes, which were exacerbated with IFNɣ exposure. Taken together, our data reveal that IFNɣ induces APOL1 expression, and that each additional RV associates with mitochondrial dysfunction and autophagy inhibition. IFNɣ amplifies this phenotype even in 1RV HUVECs, representing the first description of APOL1 pathobiology in variant heterozygous cell cultures.
Collapse
Affiliation(s)
- Ashira Blazer
- Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, NY, United States
| | - Yingzhi Qian
- Division of Biostatistics, Department of Population Health, New York University School of Medicine, New York, NY, United States
| | - Martin Paul Schlegel
- Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY, United States
| | - Huda Algasas
- Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, NY, United States
| | - Jill P. Buyon
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Ken Cadwell
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, United States
| | - Michael Cammer
- DART Microscopy Laboratory, New York University Grossman School of Medicine, New York University School of Medicine, New York, NY, United States
| | - Sean P. Heffron
- Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY, United States
| | - Feng-Xia Liang
- DART Microscopy Laboratory, New York University Grossman School of Medicine, New York University School of Medicine, New York, NY, United States
| | - Shilpi Mehta-Lee
- Department of Obstetrics and Gynecology, New York University Grossman School of Medicine, New York, NY, United States
| | - Timothy Niewold
- Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, NY, United States
| | - Sara E. Rasmussen
- Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, NY, United States
| | - Robert M. Clancy
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
17
|
Liu T, Jin Q, Ren F, Yang L, Mao H, Ma F, Wang Y, Li P, Zhan Y. Potential therapeutic effects of natural compounds targeting autophagy to alleviate podocyte injury in glomerular diseases. Biomed Pharmacother 2022; 155:113670. [PMID: 36116248 DOI: 10.1016/j.biopha.2022.113670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 11/02/2022] Open
Abstract
Podocyte injury is a common cause of proteinuric kidney diseases. Uncontrollable progressive podocyte loss accelerates glomerulosclerosis and increases the risk of end-stage renal disease. To date, owing to the complex pathological mechanism, effective therapies for podocyte injury have been limited. Accumulating evidence supports the indispensable role of autophagy in the maintenance of podocyte homeostasis. A variety of natural compounds and their derivatives have been found to regulate autophagy through multiple targets, including promotes nuclear transfer of transcription factor EB and lysosomal repair. Here, we reviewed the recent studies on the use of natural compounds and their derivatives as autophagy regulators and discussed their potential applications in ameliorating podocyte injury. Several known natural compounds with autophagy-regulatory properties, such as quercetin, silibinin, kaempferol, and artemisinin, and their medical uses were also discussed. This review will help in improving the understanding of the podocyte protective mechanism of natural compounds and promote their development for clinical use.
Collapse
Affiliation(s)
- Tongtong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qi Jin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Feihong Ren
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liping Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huimin Mao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fang Ma
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuyang Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ping Li
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China.
| | - Yongli Zhan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
18
|
Hok-A-Hin YS, Dijkstra AA, Rábano A, Hoozemans JJ, Castillo L, Seelaar H, van Swieten JC, Pijnenburg YAL, Teunissen CE, Del Campo M. Apolipoprotein L1 is increased in frontotemporal lobar degeneration post-mortem brain but not in ante-mortem cerebrospinal fluid. Neurobiol Dis 2022; 172:105813. [PMID: 35820647 DOI: 10.1016/j.nbd.2022.105813] [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: 01/31/2022] [Revised: 06/27/2022] [Accepted: 07/06/2022] [Indexed: 10/17/2022] Open
Abstract
AIMS Frontotemporal Dementia (FTD) is caused by frontal-temporal lobar degeneration (FTLD), characterized mainly by brain protein aggregates of tau (FTLD-Tau) or TDP-43 (FTLD-TDP). The clinicopathological heterogeneity makes ante-mortem diagnosis of these pathological subtypes challenging. Our proteomics study showed increased Apolipoprotein L1 (APOL1) levels in CSF from FTD patients, which was prominently expressed in FTLD-Tau. We aimed to understand APOL1 expression in FTLD post-mortem brain tissue and to validate its potential as a CSF biomarker for FTD and its pathological subtypes. METHODS APOL1 levels were analyzed in the frontal cortex of FTLD (including FTLD-Tau and FTLD-TDP) and non-demented controls by immunohistochemistry (FTLD total = 18 (12 FTLD-Tau and 6 FTLD-TDP); controls = 9), western blot (WB), and a novel prototype ELISA (FTLD total = 44 (21 FTLD-Tau and 23 FTLD-TDP); controls = 9). The association of APOL1 immunoreactivity with phosphorylated Tau (pTau) and TDP-43 (pTDP-43) immunoreactivity was assessed. CSF APOL1 was analyzed in confirmed FTD patients (n = 27, including 12 FTLD-Tau and 15 FTLD-TDP) and controls (n = 15) using the same ELISA. RESULTS APOL1 levels were significantly increased in FTLD post-mortem tissue compared to controls as measured by immunohistochemistry, WB, and ELISA. However, no differences between the pathological subtypes were observed. APOL1 immunoreactivity was present in neuronal and glial cells but did not co-localize with pTau or pTDP-43. CSF APOL1 levels were comparable between FTD patients and controls and between pathological subtypes. CONCLUSION APOL1 is upregulated in FTLD pathology irrespective of the subtypes, indicating a role of this novel protein in FTD pathophysiology. The APOL1 levels detected in brain tissue were not mirrored in the CSF, limiting its potential as a specific FTD biofluid-based biomarker using our current immunoassay.
Collapse
Affiliation(s)
- Yanaika S Hok-A-Hin
- Neurochemistry Laboratory, Clinical Chemistry Department, Amsterdam Neuroscience, VU University Medical Centers, the Netherlands.
| | - Anke A Dijkstra
- Department of Pathology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, the Netherlands
| | - Alberto Rábano
- CIEN Tissue Bank, Alzheimer's Centre Reina Sofía-CIEN Foundation, Madrid, Spain
| | - Jeroen J Hoozemans
- Department of Pathology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, the Netherlands
| | - Lucía Castillo
- Neurochemistry Laboratory, Clinical Chemistry Department, Amsterdam Neuroscience, VU University Medical Centers, the Netherlands
| | - Harro Seelaar
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, the Netherlands
| | - John C van Swieten
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Centre Amsterdam and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, the Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Clinical Chemistry Department, Amsterdam Neuroscience, VU University Medical Centers, the Netherlands
| | - Marta Del Campo
- Neurochemistry Laboratory, Clinical Chemistry Department, Amsterdam Neuroscience, VU University Medical Centers, the Netherlands; Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo- CEU, CEU Universities, Madrid, Spain
| |
Collapse
|
19
|
McNulty MT, Fermin D, Eichinger F, Jang D, Kretzler M, Burtt NP, Pollak MR, Flannick J, Weins A, Friedman DJ, Sampson MG. A glomerular transcriptomic landscape of apolipoprotein L1 in Black patients with focal segmental glomerulosclerosis. Kidney Int 2022; 102:136-148. [PMID: 34929253 PMCID: PMC9206042 DOI: 10.1016/j.kint.2021.10.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 10/20/2021] [Accepted: 10/29/2021] [Indexed: 12/26/2022]
Abstract
Apolipoprotein L1 (APOL1)-associated focal segmental glomerulosclerosis (FSGS) is the dominant form of FSGS in Black individuals. There are no targeted therapies for this condition, in part because the molecular mechanisms underlying APOL1's pathogenic contribution to FSGS are incompletely understood. Studying the transcriptomic landscape of APOL1 FSGS in patient kidneys is an important way to discover genes and molecular behaviors that are unique or most relevant to the human disease. With the hypothesis that the pathology driven by the high-risk APOL1 genotype is reflected in alteration of gene expression across the glomerular transcriptome, we compared expression and co-expression profiles of 15,703 genes in 16 Black patients with FSGS at high-risk vs 14 Black patients with a low-risk APOL1 genotype. Expression data from APOL1-inducible HEK293 cells and normal human glomeruli were used to pursue genes and molecular pathways uncovered in these studies. We discovered increased expression of APOL1 and nine other significant differentially expressed genes in high-risk patients. This included stanniocalcin, which has a role in mitochondrial and calcium-related processes along with differential correlations between high- and low-risk APOL1 and metabolism pathway genes. There were similar correlations with extracellular matrix- and immune-related genes, but significant loss of co-expression of mitochondrial genes in high-risk FSGS, and an NF-κB-down regulating gene, NKIRAS1, as the most significant hub gene with strong differential correlations with NDUF family (mitochondrial respiratory genes) and immune-related (JAK-STAT) genes. Thus, differences in mitochondrial gene regulation appear to underlie many differences observed between high- and low-risk Black patients with FSGS.
Collapse
Affiliation(s)
- Michelle T McNulty
- Division of Pediatric Nephrology, Boston Children's Hospital, Boston, Massachusetts, USA; Kidney Disease Initiative, Broad Institute, Cambridge, Massachusetts, USA
| | - Damian Fermin
- Division of Nephrology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Felix Eichinger
- Division of Nephrology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Dongkeun Jang
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Noël P Burtt
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA; Metabolism Program, Broad Institute, Cambridge, Massachusetts, USA
| | - Martin R Pollak
- Harvard Medical School, Boston, Massachusetts, USA; Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jason Flannick
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA; Metabolism Program, Broad Institute, Cambridge, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA; Division of Genetics, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Astrid Weins
- Harvard Medical School, Boston, Massachusetts, USA
| | - David J Friedman
- Harvard Medical School, Boston, Massachusetts, USA; Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew G Sampson
- Division of Pediatric Nephrology, Boston Children's Hospital, Boston, Massachusetts, USA; Kidney Disease Initiative, Broad Institute, Cambridge, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
20
|
Gerstner L, Chen M, Kampf LL, Milosavljevic J, Lang K, Schneider R, Hildebrandt F, Helmstädter M, Walz G, Hermle T. Inhibition of endoplasmic reticulum stress signaling rescues cytotoxicity of human apolipoprotein-L1 risk variants in Drosophila. Kidney Int 2022; 101:1216-1231. [PMID: 35120995 PMCID: PMC10061223 DOI: 10.1016/j.kint.2021.12.031] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 12/10/2021] [Accepted: 12/23/2021] [Indexed: 01/01/2023]
Abstract
Risk variants of the apolipoprotein-L1 (APOL1) gene are associated with severe kidney disease, putting homozygous carriers at risk. Since APOL1 lacks orthologs in all major model organisms, a wide range of mechanisms frequently in conflict have been described for APOL1-associated nephropathies. The genetic toolkit in Drosophila allows unique in vivo insights into disrupted cellular homeostasis. To perform a mechanistic analysis, we expressed human APOL1 control and gain-of-function kidney risk variants in the podocyte-like garland cells of Drosophila nephrocytes and a wing precursor tissue. Expression of APOL1 risk variants was found to elevate endocytic function of garland cell nephrocytes that simultaneously showed early signs of cell death. Wild-type APOL1 had a significantly milder effect, while a control transgene with deletion of the short BH3 domain showed no overt phenotype. Nephrocyte endo-lysosomal function and slit diaphragm architecture remained unaffected by APOL1 risk variants, but endoplasmic reticulum (ER) swelling, chaperone induction, and expression of the reporter Xbp1-EGFP suggested an ER stress response. Pharmacological inhibition of ER stress diminished APOL1-mediated cell death and direct ER stress induction enhanced nephrocyte endocytic function similar to expression of APOL1 risk variants. We confirmed APOL1-dependent ER stress in the Drosophila wing precursor where silencing the IRE1-dependent branch of ER stress signaling by inhibition with Xbp1-RNAi abrogated cell death, representing the first rescue of APOL1-associated cytotoxicity in vivo. Thus, we uncovered ER stress as an essential consequence of APOL1 risk variant expression in vivo in Drosophila, suggesting a central role of this pathway in the pathogenesis of APOL1-associated nephropathies.
Collapse
Affiliation(s)
- Lea Gerstner
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Mengmeng Chen
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Lina L Kampf
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Julian Milosavljevic
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Konrad Lang
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Ronen Schneider
- Renal Division, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Friedhelm Hildebrandt
- Renal Division, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Martin Helmstädter
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Gerd Walz
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Tobias Hermle
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany.
| |
Collapse
|
21
|
Deng J, Zhang Q, Lv L, Ma P, Zhang Y, Zhao N, Zhang Y. Identification of an autophagy-related gene signature for predicting prognosis and immune activity in pancreatic adenocarcinoma. Sci Rep 2022; 12:7006. [PMID: 35488119 PMCID: PMC9054801 DOI: 10.1038/s41598-022-11050-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 03/10/2022] [Indexed: 12/11/2022] Open
Abstract
Adenocarcinoma of the pancreas (PAAD) is a cancerous growth that deteriorates rapidly and has a poor prognosis. Researchers are investigating autophagy in PAAD to identify a new biomarker and treatment target. An autophagy-related gene (ARG) model for overall survival (OS) was constructed using multivariate Cox regression analyses. A cohort of the Cancer Genome Atlas (TCGA)-PAAD was used as the training group as a basis for model construction. This prediction model was validated with several external datasets. To evaluate model performance, the analysis with receiver operating characteristic curves (ROC) was performed. The Human Protein Atlas (HPA) and Cancer Cell Line Encyclopedia (CCLE) were investigated to validate the effects of ARGs expression on cancer cells. Comparing the levels of immune infiltration between high-risk and low-risk groups was finished through the use of CIBERSORT. The differentially expressed genes (DEGs) between the low-/high-risk groups were analyzed further via Gene Ontology biological process (GO-BP) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, which were used to identify potential small-molecule compounds in Connectivity Map (CMap), followed by half-maximal inhibitory concentration (IC50) examination with PANC-1 cells. The risk score was finally calculated as follows: BAK1 × 0.34 + ITGA3 × 0.38 + BAG3 × 0.35 + APOL1 × 0.26-RAB24 × 0.67519. ITGA3 and RAB24 both emerged as independent prognostic factors in multivariate Cox regression. Each PAAD cohort had a significantly shorter OS in the high-risk group than in the low-risk group. The high-risk group exhibited infiltration of several immune cell types, including naive B cells (p = 0.003), plasma cells (p = 0.044), and CD8 T cells (nearly significant, p = 0.080). Higher infiltration levels of NK cells (p = 0.025), resting macrophages (p = 0.020), and mast cells (p = 0.007) were found in the high-risk group than the low-risk group. The in vitro and in vivo expression of signature ARGs was consistent in the CCLE and HPA databases. The top 3 enriched Gene Ontology biological processes (GO-BPs) were signal release, regulation of transsynaptic signaling, and modulation of chemical synaptic transmission, and the top 3 enriched Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were MAPK, cAMP, and cell adhesion molecules. Four potential small-molecule compounds (piperacetazine, vinburnine, withaferin A and hecogenin) that target ARGs were also identified. Taking the results together, our research shows that the ARG signature may serve as a useful prognostic indicator and reveal potential therapeutic targets in patients with PAAD.
Collapse
Affiliation(s)
- Jiang Deng
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, People's Republic of China
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, People's Republic of China
| | - Qian Zhang
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, People's Republic of China
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, People's Republic of China
| | - Liping Lv
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, People's Republic of China
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, People's Republic of China
| | - Ping Ma
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, People's Republic of China
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, People's Republic of China
| | - Yangyang Zhang
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, People's Republic of China
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, People's Republic of China
| | - Ning Zhao
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, People's Republic of China
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, People's Republic of China
| | - Yanyu Zhang
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, People's Republic of China.
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, People's Republic of China.
| |
Collapse
|
22
|
Prescott L. SARS-CoV-2 3CLpro whole human proteome cleavage prediction and enrichment/depletion analysis. Comput Biol Chem 2022; 98:107671. [PMID: 35429835 PMCID: PMC8958254 DOI: 10.1016/j.compbiolchem.2022.107671] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 03/21/2022] [Accepted: 03/25/2022] [Indexed: 12/12/2022]
Abstract
A novel coronavirus (SARS-CoV-2) has devastated the globe as a pandemic that has killed millions of people. Widespread vaccination is still uncertain, so many scientific efforts have been directed toward discovering antiviral treatments. Many drugs are being investigated to inhibit the coronavirus main protease, 3CLpro, from cleaving its viral polyprotein, but few publications have addressed this protease’s interactions with the host proteome or their probable contribution to virulence. Too few host protein cleavages have been experimentally verified to fully understand 3CLpro’s global effects on relevant cellular pathways and tissues. Here, I set out to determine this protease’s targets and corresponding potential drug targets. Using a neural network trained on cleavages from 392 coronavirus proteomes with a Matthews correlation coefficient of 0.985, I predict that a large proportion of the human proteome is vulnerable to 3CLpro, with 4898 out of approximately 20,000 human proteins containing at least one putative cleavage site. These cleavages are nonrandomly distributed and are enriched in the epithelium along the respiratory tract, brain, testis, plasma, and immune tissues and depleted in olfactory and gustatory receptors despite the prevalence of anosmia and ageusia in COVID-19 patients. Affected cellular pathways include cytoskeleton/motor/cell adhesion proteins, nuclear condensation and other epigenetics, host transcription and RNAi, ribosomal stoichiometry and nascent-chain detection and degradation, ubiquitination, pattern recognition receptors, coagulation, lipoproteins, redox, and apoptosis. This whole proteome cleavage prediction demonstrates the importance of 3CLpro in expected and nontrivial pathways affecting virulence, lead me to propose more than a dozen potential therapeutic targets against coronaviruses, and should therefore be applied to all viral proteases and subsequently experimentally verified.
Collapse
|
23
|
Comparative Analysis of the APOL1 Variants in the Genetic Landscape of Renal Carcinoma Cells. Cancers (Basel) 2022; 14:cancers14030733. [PMID: 35159001 PMCID: PMC8833631 DOI: 10.3390/cancers14030733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/11/2022] [Accepted: 01/26/2022] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Renal cell carcinoma (RCC) occurs at higher frequency in individuals of African ancestry, with well-recorded documentation in this community. This is most prominent in the context of chronic kidney disease. In turn, many forms of progressive chronic kidney disease are more common in populations of Sub-Saharan African ancestry. This disparity has been attributed to well-defined allelic variants and has risen in the parental populations to high frequency under evolutionary pressure. Mechanisms of increased kidney disease risk and cell injury, causally associated with these APOL1 gene variants, have been extensively studied. Most studies have compared the effects of ectopic overexpression of the parental non-risk APOL1 with the mutated risk variants in cellular and organismal platforms. In the current study, we have used CRISPR/Cas9 genetic engineering to knock out or modify the sequence of endogenous APOL1 in RCC to mimic and examine the effects of these naturally occurring kidney disease risk allelic variants. Remarkably, these modifications to endogenous APOL1 genes in RCC resulted in a set of prominent effects on mitochondrial integrity and metabolic pathways and disrupted tumorigenesis. These findings both clarify pathways of cell injury of APOL1 risk variants in cells of kidney origin and motivate further studies to examine the potential central role of APOL1 in the pathogenesis of renal cell carcinoma and its relation to chronic kidney disease in genotypically at-risk African ancestry individuals. Abstract Although the relative risk of renal cell carcinoma associated with chronic kidney injury is particularly high among sub-Saharan African ancestry populations, it is unclear yet whether the APOL1 gene risk variants (RV) for kidney disease additionally elevate this risk. APOL1 G1 and G2 RV contribute to increased risk for kidney disease in black populations, although the disease mechanism has still not been fully deciphered. While high expression levels of all three APOL1 allelic variants, G0 (the wild type allele), G1, and G2 are injurious to normal human cells, renal carcinoma cells (RCC) naturally tolerate inherent high expression levels of APOL1. We utilized CRISPR/Cas9 gene editing to generate isogenic RCC clones expressing APOL1 G1 or G2 risk variants on a similar genetic background, thus enabling a reliable comparison between the phenotypes elicited in RCC by each of the APOL1 variants. Here, we demonstrate that knocking in the G1 or G2 APOL1 alleles, or complete elimination of APOL1 expression, has major effects on proliferation capacity, mitochondrial morphology, cell metabolism, autophagy levels, and the tumorigenic potential of RCC cells. The most striking effect of the APOL1 RV effect was demonstrated in vivo by the complete abolishment of tumor growth in immunodeficient mice. Our findings suggest that, in contrast to the WT APOL1 variant, APOL1 RV are toxic for RCC cells and may act to suppress cancer cell growth. We conclude that the inherent expression of non-risk APOL1 G0 is required for RCC tumorigenicity. RCC cancer cells can hardly tolerate increased APOL1 risk variants expression levels as opposed to APOL1 G0.
Collapse
|
24
|
Identification of a Prognosis-Related Risk Signature for Bladder Cancer to Predict Survival and Immune Landscapes. J Immunol Res 2021; 2021:3236384. [PMID: 34708131 PMCID: PMC8545590 DOI: 10.1155/2021/3236384] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/14/2021] [Accepted: 09/22/2021] [Indexed: 12/22/2022] Open
Abstract
Background Bladder cancer is the tenth most common cancer worldwide. Valuable biomarkers in the field of diagnostic bladder cancer are urgently required. Method Here, the gene expression matrix and clinical data were obtained from The Cancer Genome Atlas (TCGA), GSE13507, GSE32894, and Mariathasan et al. Five prognostic genes were identified by the univariate, robust, and multivariate Cox's regression and were used to develop a prognosis-related model. The Kaplan-Meier survival curves and receiver operating characteristics were used to evaluate the model's effectiveness. The potential biological functions of the selected genes were analyzed using CIBERSORT and ESTIMATE algorithms. Cancer Therapeutics Response Portal (CTRP) and PRISM datasets were used to identify drugs with high sensitivity. Subsequently, using the bladder cancer (BLCA) cell lines, the role of TNFRSF14 was determined by Western blotting, cell proliferation assay, and 5-ethynyl-20-deoxyuridine assay. Results GSDMB, CLEC2D, APOL2, TNFRSF14, and GBP2 were selected as prognostic genes in bladder cancer patients. The model's irreplaceable reliability was validated by the training and validation cohorts. CD8+ T cells were highly infiltrated in the high-TNFRSF14-expression group, and M2 macrophages were the opposite. Higher expression of TNFRSF14 was associated with higher expression levels of LCK, interferon, MHC-I, and MHC-II, while risk score was the opposite. Many compounds with higher sensitivity for treating bladder cancer patients in the low-TNFRSF14-expression group were identified, with obatoclax being a potential drug most likely to treat patients in the low-TNFRSF14-expression group. Finally, the proliferation of BLCA cell lines was increased in the TNFRSF14-reduced group, and the differential expression was identified. TNFRSF14 plays a role in bladder cancer progression through the Wnt/β-catenin-dependent pathway. TNFRSF14 is a potential protective biomarker involved in cell proliferation in BLCA. Conclusion We conducted a study to establish a 5-gene score model, providing reliable prediction for the outcome of bladder cancer patients and therapeutic drugs to individualize therapy. Our findings provide a signature that might help determine the optimal treatment for individual patients with bladder cancer.
Collapse
|
25
|
Müller D, Schmitz J, Fischer K, Granado D, Groh AC, Krausel V, Lüttgenau SM, Amelung TM, Pavenstädt H, Weide T. Evolution of Renal-Disease Factor APOL1 Results in Cis and Trans Orientations at the Endoplasmic Reticulum That Both Show Cytotoxic Effects. Mol Biol Evol 2021; 38:4962-4976. [PMID: 34323996 PMCID: PMC8557400 DOI: 10.1093/molbev/msab220] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The recent and exclusively in humans and a few other higher primates expressed APOL1 (apolipoprotein L1) gene is linked to African human trypanosomiasis (also known as African sleeping sickness) as well as to different forms of kidney diseases. Whereas APOL1's role as a trypanolytic factor is well established, pathobiological mechanisms explaining its cytotoxicity in renal cells remain unclear. In this study, we compared the APOL family members using a combination of evolutionary studies and cell biological experiments to detect unique features causal for APOL1 nephrotoxic effects. We investigated available primate and mouse genome and transcriptome data to apply comparative phylogenetic and maximum likelihood selection analyses. We suggest that the APOL gene family evolved early in vertebrates and initial splitting occurred in ancestral mammals. Diversification and differentiation of functional domains continued in primates, including developing the two members APOL1 and APOL2. Their close relationship could be diagnosed by sequence similarity and a shared ancestral insertion of an AluY transposable element. Live-cell imaging analyses showed that both expressed proteins show a strong preference to localize at the endoplasmic reticulum (ER). However, glycosylation and secretion assays revealed that-unlike APOL2-APOL1 membrane insertion or association occurs in different orientations at the ER, with the disease-associated mutants facing either the luminal (cis) or cytoplasmic (trans) side of the ER. The various pools of APOL1 at the ER offer a novel perspective in explaining the broad spectrum of its observed toxic effects.
Collapse
Affiliation(s)
- Daria Müller
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Jürgen Schmitz
- Institute of Experimental Pathology, ZMBE, University of Münster, Münster, Germany
| | - Katharina Fischer
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Daniel Granado
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Ann-Christin Groh
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Vanessa Krausel
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Simona Mareike Lüttgenau
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Till Maximilian Amelung
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Hermann Pavenstädt
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Thomas Weide
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| |
Collapse
|
26
|
Hu J, Stojanović J, Yasamineh S, Yasamineh P, Karuppannan SK, Hussain Dowlath MJ, Serati-Nouri H. The potential use of microRNAs as a therapeutic strategy for SARS-CoV-2 infection. Arch Virol 2021; 166:2649-2672. [PMID: 34278528 PMCID: PMC8286877 DOI: 10.1007/s00705-021-05152-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/21/2021] [Indexed: 02/06/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). To date, there is no effective therapeutic approach for treating SARS-CoV-2 infections. MicroRNAs (miRNAs) have been recognized to target the viral genome directly or indirectly, thereby inhibiting viral replication. Several studies have demonstrated that host miRNAs target different sites in SARS-CoV-2 RNA and constrain the production of essential viral proteins. Furthermore, miRNAs have lower toxicity, are more immunogenic, and are more diverse than protein-based and even plasmid-DNA-based therapeutic agents. In this review, we emphasize the role of miRNAs in viral infection and their potential use as therapeutic agents against COVID-19 disease. The potential of novel miRNA delivery strategies, especially EDV™ nanocells, for targeting lung tissue for treatment of SARS-CoV-2 infection is also discussed.
Collapse
Affiliation(s)
- Jiulue Hu
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, China
| | - Jelena Stojanović
- Faculty of Mathematics and Computer Science in Belgrade, ALFA BK University, Belgrade, Serbia
| | - Saman Yasamineh
- Young Researcher and Elite Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran.
| | - Pooneh Yasamineh
- Young Researcher and Elite Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Sathish Kumar Karuppannan
- Center for Environmental Nuclear Research, Directorate of Research and Virtual Education, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603203, Kanchipuram, Chennai, Tamil Nadu, India
| | - Mohammed Junaid Hussain Dowlath
- Center for Environmental Nuclear Research, Directorate of Research and Virtual Education, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603203, Kanchipuram, Chennai, Tamil Nadu, India
| | - Hamed Serati-Nouri
- Stem cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
27
|
Hashemi SMA, Thijssen M, Hosseini SY, Tabarraei A, Pourkarim MR, Sarvari J. Human gene polymorphisms and their possible impact on the clinical outcome of SARS-CoV-2 infection. Arch Virol 2021; 166:2089-2108. [PMID: 33934196 PMCID: PMC8088757 DOI: 10.1007/s00705-021-05070-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 02/23/2021] [Indexed: 12/13/2022]
Abstract
The SARS-CoV-2 pandemic has become one of the most serious health concerns globally. Although multiple vaccines have recently been approved for the prevention of coronavirus disease 2019 (COVID-19), an effective treatment is still lacking. Our knowledge of the pathogenicity of this virus is still incomplete. Studies have revealed that viral factors such as the viral load, duration of exposure to the virus, and viral mutations are important variables in COVID-19 outcome. Furthermore, host factors, including age, health condition, co-morbidities, and genetic background, might also be involved in clinical manifestations and infection outcome. This review focuses on the importance of variations in the host genetic background and pathogenesis of SARS-CoV-2. We will discuss the significance of polymorphisms in the ACE-2, TMPRSS2, vitamin D receptor, vitamin D binding protein, CD147, glucose-regulated protein 78 kDa, dipeptidyl peptidase-4 (DPP4), neuropilin-1, heme oxygenase, apolipoprotein L1, vitamin K epoxide reductase complex 1 (VKORC1), and immune system genes for the clinical outcome of COVID-19.
Collapse
Affiliation(s)
- Seyed Mohammad Ali Hashemi
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Microbiology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Marijn Thijssen
- Laboratory for Clinical and Epidemiological Virology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, 3000 Leuven, Belgium
| | - Seyed Younes Hosseini
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alijan Tabarraei
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mahmoud Reza Pourkarim
- Laboratory for Clinical and Epidemiological Virology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, 3000 Leuven, Belgium
- Health Policy Research Centre, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jamal Sarvari
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
28
|
Ultsch M, Holliday MJ, Gerhardy S, Moran P, Scales SJ, Gupta N, Oltrabella F, Chiu C, Fairbrother W, Eigenbrot C, Kirchhofer D. Structures of the ApoL1 and ApoL2 N-terminal domains reveal a non-classical four-helix bundle motif. Commun Biol 2021; 4:916. [PMID: 34316015 PMCID: PMC8316464 DOI: 10.1038/s42003-021-02387-5] [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] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/23/2021] [Indexed: 02/07/2023] Open
Abstract
Apolipoprotein L1 (ApoL1) is a circulating innate immunity protein protecting against trypanosome infection. However, two ApoL1 coding variants are associated with a highly increased risk of chronic kidney disease. Here we present X-ray and NMR structures of the N-terminal domain (NTD) of ApoL1 and of its closest relative ApoL2. In both proteins, four of the five NTD helices form a four-helix core structure which is different from the classical four-helix bundle and from the pore-forming domain of colicin A. The reactivity with a conformation-specific antibody and structural models predict that this four-helix motif is also present in the NTDs of ApoL3 and ApoL4, suggesting related functions within the small ApoL family. The long helix 5 of ApoL1 is conformationally flexible and contains the BH3-like region. This BH3-like α-helix resembles true BH3 domains only in sequence and structure but not in function, since it does not bind to the pro-survival members of the Bcl-2 family, suggesting a Bcl-2-independent role in cytotoxicity. These findings should expedite a more comprehensive structural and functional understanding of the ApoL immune protein family.
Collapse
Affiliation(s)
- Mark Ultsch
- Department of Structural Biology, Genentech Inc., South San Francisco, CA, USA
| | - Michael J Holliday
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA, USA
| | - Stefan Gerhardy
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA, USA
| | - Paul Moran
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA, USA
| | - Suzie J Scales
- Department of Immunology, Genentech Inc., South San Francisco, CA, USA
| | - Nidhi Gupta
- Department of Immunology, Genentech Inc., South San Francisco, CA, USA
| | | | - Cecilia Chiu
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| | - Wayne Fairbrother
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA, USA
| | - Charles Eigenbrot
- Department of Structural Biology, Genentech Inc., South San Francisco, CA, USA
| | - Daniel Kirchhofer
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA, USA.
| |
Collapse
|
29
|
High-Density Lipoproteins and the Kidney. Cells 2021; 10:cells10040764. [PMID: 33807271 PMCID: PMC8065870 DOI: 10.3390/cells10040764] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/28/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
Dyslipidemia is a typical trait of patients with chronic kidney disease (CKD) and it is typically characterized by reduced high-density lipoprotein (HDL)-cholesterol(c) levels. The low HDL-c concentration is the only lipid alteration associated with the progression of renal disease in mild-to-moderate CKD patients. Plasma HDL levels are not only reduced but also characterized by alterations in composition and structure, which are responsible for the loss of atheroprotective functions, like the ability to promote cholesterol efflux from peripheral cells and antioxidant and anti-inflammatory proprieties. The interconnection between HDL and renal function is confirmed by the fact that genetic HDL defects can lead to kidney disease; in fact, mutations in apoA-I, apoE, apoL, and lecithin–cholesterol acyltransferase (LCAT) are associated with the development of renal damage. Genetic LCAT deficiency is the most emblematic case and represents a unique tool to evaluate the impact of alterations in the HDL system on the progression of renal disease. Lipid abnormalities detected in LCAT-deficient carriers mirror the ones observed in CKD patients, which indeed present an acquired LCAT deficiency. In this context, circulating LCAT levels predict CKD progression in individuals at early stages of renal dysfunction and in the general population. This review summarizes the main alterations of HDL in CKD, focusing on the latest update of acquired and genetic LCAT defects associated with the progression of renal disease.
Collapse
|
30
|
Sun B, Zhai S, Zhang L, Sun G. The role of extracellular vesicles in podocyte autophagy in kidney disease. J Cell Commun Signal 2021; 15:299-316. [PMID: 33619681 DOI: 10.1007/s12079-020-00594-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
Podocytes are the key cells involved in protein filtration in the glomerulus. Once proteins appear in the urine when podocytes fail, patients will end with renal failure due to the progression of glomerular damage if no proper treatment is applied. The injury and loss of podocytes can be attributed to diverse factors, such as genetic, immunologic, toxic, or metabolic disorders. Recently, autophagy has emerged as a key mechanism to eliminate the unwanted cytoplasmic materials and to prolong the lifespan of podocytes by alleviating cell damage and stress. Typically, the fundamental function of extracellular vesicles (EVs) is to mediate the intercellular communication. Recent studies have suggested that, EVs, especially exosomes, play a certain role in information transfer by communicating proteins, mRNAs, and microRNAs with recipient cells. Under physiological and pathological conditions, EVs assist in the bioinformation interchange between kidneys and other organs. It is suggested that EVs are related to the pathogenesis of acute kidney injury and chronic kidney disease, including glomerular disease, diabetic nephropathy, renal fibrosis and end-stage renal disease. However, the role of EVs in podocyte autophagy remains unclear so far. Here, this study integrated the existing information about the relevancy, diagnostic value and therapeutic potential of EVs in a variety of podocytes-related diseases. The accumulating evidence highlighted that autophagy played a critical role in the homeostasis of podocytes in glomerular disease.
Collapse
Affiliation(s)
- Baichao Sun
- Department of Nephrology, The Second Hospital of Jilin University, 218 ZiQiang Street, Changchun, 130041, Jilin, People's Republic of China.,Department of Pediatric Nephrology, The First Hospital of Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Shubo Zhai
- Department of Pediatric Nephrology, The First Hospital of Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Li Zhang
- Department of Pediatric Nephrology, The First Hospital of Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Guangdong Sun
- Department of Nephrology, The Second Hospital of Jilin University, 218 ZiQiang Street, Changchun, 130041, Jilin, People's Republic of China.
| |
Collapse
|
31
|
Zou D, Wang Y, Wang M, Zhao B, Hu F, Li Y, Zhang B. Bioinformatics analysis reveals the competing endogenous RNA (ceRNA) coexpression network in the tumor microenvironment and prognostic biomarkers in soft tissue sarcomas. Bioengineered 2021; 12:496-506. [PMID: 33587010 PMCID: PMC8806339 DOI: 10.1080/21655979.2021.1879566] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Soft tissue sarcomas (STSs) are rare, heterogeneous mesenchymal neoplasias. Understanding the tumor microenvironment (TME) and identifying potential biomarkers for prognosis associated with the TME of STS might provide effective clues for immune therapy. We evaluated the immune scores and stromal scores of STS patients by using the RNA sequencing dataset from The Cancer Genome Atlas (TCGA) database and the ESTIMATE algorithm. Then, the differentially expressed mRNAs (DEGs), miRNAs (DEMs) and lncRNAs (DELs) were identified after comparing the high- and low-score groups. Next, we established a competing endogenous RNA (ceRNA) network and explored the prognostic values of biomarkers involved in the network with the help of bioinformatics analysis. High immune score was significantly associated with favorable overall survival in STS patients. A total of 328 DEGs, 18 DEMs and 67 DELs commonly regulated in the immune and stromal score groups were obtained. A ceRNA network and protein-protein interaction (PPI) network identified some hub nodes with considerable importance in the network. Kaplan-Meier survival analysis demonstrated that nine mRNAs, two miRNAs and three lncRNAs were closely associated with overall survival of STS patients. Gene set enrichment analysis (GSEA) suggested that these three lncRNAs were mainly involved in immune response-associated pathways in STS patients. Finally, the expression levels of five mRNAs (APOL1, EFEMP1, LYZ, RARRES1 and TNFAIP2) were verified, which were consistent with the results of the TCGA cohort. The results of our study confirmed the prognostic value of immune scores for STS patients. We also identified several TME-related biomarkers that might contribute to prognostic prediction and immune therapy.
Collapse
Affiliation(s)
- Dandan Zou
- Department of Radiology, The First Hospital of Qinhuangdao , Qinhuangdao, Hebei, China
| | - Yang Wang
- Department of MRI, The Third Hospital of Qinhuangdao , Qinhuangdao, Hebei, China
| | - Meng Wang
- Department of Clinical Laboratory, The First Hospital of Qinhuangdao , Qinhuangdao, Hebei, China
| | - Bo Zhao
- Department of Radiology, The First Hospital of Qinhuangdao , Qinhuangdao, Hebei, China
| | - Fei Hu
- Department of Radiology, The First Hospital of Qinhuangdao , Qinhuangdao, Hebei, China
| | - Yanguo Li
- Department of Radiology, The First Hospital of Qinhuangdao , Qinhuangdao, Hebei, China
| | - Bingming Zhang
- Department of Radiology, The First Hospital of Qinhuangdao , Qinhuangdao, Hebei, China
| |
Collapse
|
32
|
Ge M, Molina J, Ducasa GM, Mallela SK, Varona Santos J, Mitrofanova A, Kim JJ, Liu X, Sloan A, Mendez AJ, Banerjee S, Liu S, Szeto HH, Shin MK, Hoek M, Kopp JB, Fontanesi F, Merscher S, Fornoni A. APOL1 risk variants affect podocyte lipid homeostasis and energy production in focal segmental glomerulosclerosis. Hum Mol Genet 2021; 30:182-197. [PMID: 33517446 DOI: 10.1093/hmg/ddab022] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/09/2020] [Accepted: 01/12/2021] [Indexed: 12/14/2022] Open
Abstract
Lipotoxicity was recently reported in several forms of kidney disease, including focal segmental glomerulosclerosis (FSGS). Susceptibility to FSGS in African Americans is associated with the presence of genetic variants of the Apolipoprotein L1 gene (APOL1) named G1 and G2. If and how endogenous APOL1 may alter mitochondrial function by the modifying cellular lipid metabolism is unknown. Using transgenic mice expressing the APOL1 variants (G0, G1 or G2) under endogenous promoter, we show that APOL1 risk variant expression in transgenic mice does not impair kidney function at baseline. However, APOL1 G1 expression worsens proteinuria and kidney function in mice characterized by the podocyte inducible expression of nuclear factor of activated T-cells (NFAT), which we have found to cause FSGS. APOL1 G1 expression in this FSGS-model also results in increased triglyceride and cholesterol ester contents in kidney cortices, where lipid accumulation correlated with loss of renal function. In vitro, we show that the expression of endogenous APOL1 G1/G2 in human urinary podocytes is associated with increased cellular triglyceride content and is accompanied by mitochondrial dysfunction in the presence of compensatory oxidative phosphorylation (OXPHOS) complexes elevation. Our findings indicate that APOL1 risk variant expression increases the susceptibility to lipid-dependent podocyte injury, ultimately leading to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Mengyuan Ge
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Judith Molina
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - G Michelle Ducasa
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Shamroop K Mallela
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Javier Varona Santos
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Alla Mitrofanova
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Jin-Ju Kim
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Xiaochen Liu
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Alexis Sloan
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Armando J Mendez
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Santanu Banerjee
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Shaoyi Liu
- Social Profit Network Research Lab, Alexandria Launch Labs, New York, New York 10016, USA
| | - Hazel H Szeto
- Social Profit Network Research Lab, Alexandria Launch Labs, New York, New York 10016, USA
| | - Myung K Shin
- Merck & Company, Inc., Kennilworth, New Jersey 07033, USA
| | - Maarten Hoek
- Merck & Company, Inc., Kennilworth, New Jersey 07033, USA
| | - Jeffrey B Kopp
- Kidney Disease Section, NIDDK, NIH, Bethesda, Maryland 20892, USA
| | - Flavia Fontanesi
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| |
Collapse
|
33
|
Bruno J, Edwards JC. Kidney-disease-associated variants of Apolipoprotein L1 show gain of function in cation channel activity. J Biol Chem 2021; 296:100238. [PMID: 33380423 PMCID: PMC7948812 DOI: 10.1074/jbc.ra120.013943] [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] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 11/08/2022] Open
Abstract
Variants in Apolipoprotein L1 (ApoL1) are known to be responsible for increased risk of some progressive kidney diseases among people of African ancestry. ApoL1 is an amphitropic protein that can insert into phospholipid membranes and confer anion- or cation-selective permeability to phospholipid membranes depending on pH. Whether these activities differ among the variants or whether they contribute to disease pathogenesis is unknown. We used assays of voltage-driven ion flux from phospholipid vesicles and of stable membrane association to assess differences among ApoL1 isoforms. There is a significant (approximately twofold) increase in the cation-selective ion permease activity of the two kidney-disease-associated variants compared with the reference protein. In contrast, we find no difference in the anion-selective permease activity at low pH among the isoforms. Compared with the reference sequence, the two disease-associated variants show increased stable association with phospholipid vesicles under conditions that support the cation permease activity, suggesting that the increased activity may be due to more efficient membrane association and insertion. There is no difference in membrane association among isoforms under optimal conditions for the anion permease activity. These data support a model in which enhanced cation permeability may contribute to the progressive kidney diseases associated with high-risk ApoL1 alleles.
Collapse
Affiliation(s)
- Jonathan Bruno
- Nephrology Division, Department of Internal Medicine, Saint Louis University, St Louis, Missouri, USA
| | - John C Edwards
- Nephrology Division, Department of Internal Medicine, Saint Louis University, St Louis, Missouri, USA.
| |
Collapse
|
34
|
Pays E. The function of apolipoproteins L (APOLs): relevance for kidney disease, neurotransmission disorders, cancer and viral infection. FEBS J 2021; 288:360-381. [PMID: 32530132 PMCID: PMC7891394 DOI: 10.1111/febs.15444] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/24/2020] [Accepted: 06/03/2020] [Indexed: 12/17/2022]
Abstract
The discovery that apolipoprotein L1 (APOL1) is the trypanolytic factor of human serum raised interest about the function of APOLs, especially following the unexpected finding that in addition to their protective action against sleeping sickness, APOL1 C-terminal variants also cause kidney disease. Based on the analysis of the structure and trypanolytic activity of APOL1, it was proposed that APOLs could function as ion channels of intracellular membranes and be involved in mechanisms triggering programmed cell death. In this review, the recent finding that APOL1 and APOL3 inversely control the synthesis of phosphatidylinositol-4-phosphate (PI(4)P) by the Golgi PI(4)-kinase IIIB (PI4KB) is commented. APOL3 promotes Ca2+ -dependent activation of PI4KB, but due to their increased interaction with APOL3, APOL1 C-terminal variants can inactivate APOL3, leading to reduction of Golgi PI(4)P synthesis. The impact of APOLs on several pathological processes that depend on Golgi PI(4)P levels is discussed. I propose that through their effect on PI4KB activity, APOLs control not only actomyosin activities related to vesicular trafficking, but also the generation and elongation of autophagosomes induced by inflammation.
Collapse
Affiliation(s)
- Etienne Pays
- Laboratory of Molecular ParasitologyIBMMUniversité Libre de BruxellesGosseliesBelgium
| |
Collapse
|
35
|
Abstract
Evolutionary processes, including mutation, migration and natural selection, have influenced the prevalence and distribution of various disorders in humans. However, despite a few well-known examples, such as the APOL1 variants - which have undergone positive genetic selection for their ability to confer resistance to Trypanosoma brucei infection but confer a higher risk of chronic kidney disease - little is known about the effects of evolutionary processes that have shaped genetic variation on kidney disease. An understanding of basic concepts in evolutionary genetics provides an opportunity to consider how findings from ancient and archaic genomes could inform our knowledge of evolution and provide insights into how population migration and genetic admixture have shaped the current distribution and landscape of human kidney-associated diseases. Differences in exposures to infectious agents, environmental toxins, dietary components and climate also have the potential to influence the evolutionary genetics of kidneys. Of note, selective pressure on loci associated with kidney disease is often from non-kidney diseases, and thus it is important to understand how the link between genome-wide selected loci and kidney disease occurs in relation to secondary nephropathies.
Collapse
|
36
|
Privratsky JR, Li YJ, Haynes C, Podgoreanu M, Mathew J, Shah SH, Stafford-Smith M. Apolipoprotein L1 (APOL1) Coding Variants Are Associated With Creatinine Rise After Cardiac Surgery. J Cardiothorac Vasc Anesth 2020; 34:3314-3320. [PMID: 32434723 PMCID: PMC7655610 DOI: 10.1053/j.jvca.2020.04.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Acute kidney injury (AKI) is a complication of cardiac surgery that is considerably more common in African Americans (1.5-fold). Although homozygous status for apolipoprotein L1 (APOL1) risk alleles is associated with chronic kidney disease in individuals of African ancestry, whether these coding variants confer AKI risk is unknown. The present study examined whether APOL1 homozygous risk allele status was associated with AKI in African Americans after cardiac surgery. DESIGN Retrospective analysis of a cohort. SETTING Single-center university hospital. PARTICIPANTS African American patients from the CATHeterization GENetics study cohort who underwent cardiac surgery with cardiopulmonary bypass. INTERVENTIONS Genotyping of APOL1 alleles. MEASUREMENTS AND MAIN RESULTS Data from 125 African American patients included 12 APOL1 risk (ie, homozygous for risk alleles) patients and 113 APOL1 control (ie, wildtype or heterozygous for risk alleles) patients. The primary outcome to reflect AKI was peak serum creatinine rise after surgery relative to the preoperative creatinine (%ΔCr). The secondary outcome was Kidney Disease: Improving Global Outcomes (KDIGO) AKI criteria. In the primary analysis, peak creatinine rise was higher in risk compared with control patients in both univariate (%ΔCr 69.1 v 29.6%; p = 0.005) and multivariate regression (%ΔCr 88.5 v 43.7%; p = 0.006) analyses. For the secondary outcome, a trend toward KDIGO AKI development was noted in APOL1 risk patients, but this was not statistically significant. CONCLUSIONS African American cardiac surgery patients homozygous for APOL1 chronic kidney disease risk variants averaged a more than 2-fold higher postoperative creatinine rise even after adjustment for other risk factors, suggesting these alleles also are independent risk factors for AKI.
Collapse
Affiliation(s)
- Jamie R Privratsky
- Department of Anesthesiology, Duke University Medical Center, Durham, NC.
| | - Yi-Ju Li
- Department of Anesthesiology, Duke University Medical Center, Durham, NC; Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC; Duke Molecular Physiology Institute, Duke University, Durham, NC
| | - Carol Haynes
- Department of Medicine, Duke University Medical Center, Durham, NC
| | - Mihai Podgoreanu
- Department of Anesthesiology, Duke University Medical Center, Durham, NC
| | - Joseph Mathew
- Department of Anesthesiology, Duke University Medical Center, Durham, NC
| | - Svati H Shah
- Duke Molecular Physiology Institute, Duke University, Durham, NC; Department of Medicine, Duke University Medical Center, Durham, NC
| | | |
Collapse
|
37
|
Madhavan SM, Buck M. The Relationship between APOL1 Structure and Function: Clinical Implications. KIDNEY360 2020; 2:134-140. [PMID: 35368828 PMCID: PMC8785724 DOI: 10.34067/kid.0002482020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 11/04/2020] [Indexed: 02/04/2023]
Abstract
Common variants in the APOL1 gene are associated with an increased risk of nondiabetic kidney disease in individuals of African ancestry. Mechanisms by which APOL1 variants mediate kidney disease pathogenesis are not well understood. Amino acid changes resulting from the kidney disease-associated APOL1 variants alter the three-dimensional structure and conformational dynamics of the C-terminal α-helical domain of the protein, which can rationalize the functional consequences. Understanding the three-dimensional structure of the protein, with and without the risk variants, can provide insights into the pathogenesis of kidney diseases mediated by APOL1 variants.
Collapse
Affiliation(s)
| | - Matthias Buck
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
38
|
Wang DP, Yu ZX, He ZC, Liao JF, Shen XB, Zhu PL, Chen WN, Lin X, Xu SH. Apolipoprotein L1 is transcriptionally regulated by SP1, IRF1 and IRF2 in hepatoma cells. FEBS Lett 2020; 594:3108-3121. [PMID: 32671843 DOI: 10.1002/1873-3468.13887] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/21/2020] [Accepted: 07/05/2020] [Indexed: 11/07/2022]
Abstract
Apolipoprotein L1 (APOL1) participates in lipid metabolism. Here, we investigate the mechanisms regulating APOL1 gene expression in hepatoma cells. We demonstrate that the -80-nt to +31-nt region of the APOL1 promoter, which contains one SP transcription factor binding GT box and an interferon regulatory factor (IRF) binding ISRE element, maintains the maximum activity. Mutation of the GT box and ISRE element dramatically reduces APOL1 promoter activity. EMSA and chromatin immunoprecipitation assay reveal that the transcription factors Sp1, IRF1 and IRF2 could interact with their cognate binding sites on the APOL1 promoter. Overexpression of Sp1, IRF1 and IRF2 increases promoter activity, leading to increased APOL1 mRNA and protein levels, while knockdown of Sp1, IRF1 and IRF2 has the opposite effects. These results demonstrate that the APOL1 gene could be regulated by Sp1, IRF1 and IRF2 in hepatoma cells.
Collapse
Affiliation(s)
- De-Ping Wang
- Department of Medical Intensive Care Unit, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Department of Endocrinology and Metabolism, Hongqi Hospital of MuDanJiang Medical College, Mudanjiang, China
| | - Zhao-Xi Yu
- Department of Medical Intensive Care Unit, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Zong-Cun He
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Jin-Fu Liao
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xue-Bin Shen
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Department of Cardiology, Affiliated Nanping First Hospital, Fujian Medical University, Nanping, China
| | - Peng-Li Zhu
- Department of Medical Intensive Care Unit, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Wan-Nan Chen
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xu Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Shang-Hua Xu
- Department of Cardiology, Affiliated Nanping First Hospital, Fujian Medical University, Nanping, China
| |
Collapse
|
39
|
Lecordier L, Uzureau S, Vanwalleghem G, Deleu M, Crowet JM, Barry P, Moran B, Voorheis P, Dumitru AC, Yamaryo-Botté Y, Dieu M, Tebabi P, Vanhollebeke B, Lins L, Botté CY, Alsteens D, Dufrêne Y, Pérez-Morga D, Nolan DP, Pays E. The Trypanosoma Brucei KIFC1 Kinesin Ensures the Fast Antibody Clearance Required for Parasite Infectivity. iScience 2020; 23:101476. [PMID: 32889430 PMCID: PMC7479354 DOI: 10.1016/j.isci.2020.101476] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 07/30/2020] [Accepted: 08/17/2020] [Indexed: 12/24/2022] Open
Abstract
Human innate immunity to Trypanosoma brucei involves the trypanosome C-terminal kinesin TbKIFC1, which transports internalized trypanolytic factor apolipoprotein L1 (APOL1) within the parasite. We show that TbKIFC1 preferentially associates with cholesterol-containing membranes and is indispensable for mammalian infectivity. Knockdown of TbKIFC1 did not affect trypanosome growth in vitro but rendered the parasites unable to infect mice unless antibody synthesis was compromised. Surface clearance of Variant Surface Glycoprotein (VSG)-antibody complexes was far slower in these cells, which were more susceptible to capture by macrophages. This phenotype was not due to defects in VSG expression or trafficking but to decreased VSG mobility in a less fluid, stiffer surface membrane. This change can be attributed to increased cholesterol level in the surface membrane in TbKIFC1 knockdown cells. Clearance of surface-bound antibodies by T. brucei is therefore essential for infectivity and depends on high membrane fluidity maintained by the cholesterol-trafficking activity of TbKIFC1.
Collapse
Affiliation(s)
- Laurence Lecordier
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 12, rue des professeurs Jeener et Brachet, 6041 Gosselies, Belgium
| | - Sophie Uzureau
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 12, rue des professeurs Jeener et Brachet, 6041 Gosselies, Belgium
| | - Gilles Vanwalleghem
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 12, rue des professeurs Jeener et Brachet, 6041 Gosselies, Belgium
| | - Magali Deleu
- Laboratory of Molecular Biophysics at Interface (LBMI), University of Liège-Gembloux Agro Bio Tech, 2, Passage des Déportés, 5030 Gembloux, Belgium
| | - Jean-Marc Crowet
- Laboratory of Molecular Biophysics at Interface (LBMI), University of Liège-Gembloux Agro Bio Tech, 2, Passage des Déportés, 5030 Gembloux, Belgium
| | - Paul Barry
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland
| | - Barry Moran
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland
| | - Paul Voorheis
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland
| | - Andra-Cristina Dumitru
- Louvain Institute of Biomolecular Science and Technology, Catholic University of Louvain, Croix du Sud 4-5, 1348 Louvain-la-Neuve, Belgium
| | - Yoshiki Yamaryo-Botté
- Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, 38700 La Tronche, France
| | - Marc Dieu
- MaSUN, Mass Spectrometry Facility, University of Namur, 61 Rue de Bruxelles, 5000 Namur, Belgium
| | - Patricia Tebabi
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 12, rue des professeurs Jeener et Brachet, 6041 Gosselies, Belgium
| | - Benoit Vanhollebeke
- Laboratory of Neurovascular Signaling, Université Libre de Bruxelles, 12, Rue des Profs Jeener et Brachet, 6041 Gosselies, Belgium
| | - Laurence Lins
- Laboratory of Molecular Biophysics at Interface (LBMI), University of Liège-Gembloux Agro Bio Tech, 2, Passage des Déportés, 5030 Gembloux, Belgium
| | - Cyrille Y. Botté
- Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, 38700 La Tronche, France
| | - David Alsteens
- Louvain Institute of Biomolecular Science and Technology, Catholic University of Louvain, Croix du Sud 4-5, 1348 Louvain-la-Neuve, Belgium
| | - Yves Dufrêne
- Louvain Institute of Biomolecular Science and Technology, Catholic University of Louvain, Croix du Sud 4-5, 1348 Louvain-la-Neuve, Belgium
| | - David Pérez-Morga
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 12, rue des professeurs Jeener et Brachet, 6041 Gosselies, Belgium
- Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles, 12, Rue des Profs Jeener et Brachet, 6041 Gosselies, Belgium
| | - Derek P. Nolan
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland
| | - Etienne Pays
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 12, rue des professeurs Jeener et Brachet, 6041 Gosselies, Belgium
| |
Collapse
|
40
|
Valsecchi M, Cazzetta V, Oriolo F, Lan X, Piazza R, Saleem MA, Singhal PC, Mavilio D, Mikulak J, Aureli M. APOL1 polymorphism modulates sphingolipid profile of human podocytes. Glycoconj J 2020; 37:729-744. [PMID: 32915357 PMCID: PMC7679335 DOI: 10.1007/s10719-020-09944-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/19/2020] [Accepted: 09/02/2020] [Indexed: 12/01/2022]
Abstract
Apolipoprotein L1 (APOL1) wild type (G0) plays a role in the metabolism of sphingolipids, glycosphingolipids, sphingomyelin and ceramide, which constitute bioactive components of the lipid rafts (DRM). We asked whether APOL1 variants (APOL1-Vs) G1 and G2 carry the potential to alter the metabolism of sphingolipids in human podocytes. The sphingolipid pattern in HPs overexpressing either APOL1G0 or APOL1-Vs was analysed by using a thin mono- and bi-dimensional layer chromatography, mass-spectrometry and metabolic labelling with [1-3H]sphingosine. HP G0 and G1/G2-Vs exhibit a comparable decrease in lactosylceramide and an increase in the globotriaosylceramide content. An analysis of the main glycohydrolases activity involved in glycosphingolipid catabolism showed an overall decrease in the activeness of the tested enzymes, irrespective of the type of APOL1-Vs expression. Similarly, the high throughput cell live-based assay showed a comparable increased action of the plasma membrane glycosphingolipid-glycohydrolases in living cells independent of the genetic APOL1 expression profile. Importantly, the most significative modification of the sphingolipid pattern induced by APOL1-Vs occurred in DRM resulted with a drastic reduction of radioactivity associated with sphingolipids. G1/G2-Vs present a decrease amount of globotriaosylceramide and globopentaosylceramide compared to G0. Additionally, ceramide at the DRM site and lactosylceramide in general, showed a greatest fall in G1/G2 in comparison with G0. Additionally, the levels of glucosylceramide decreased only in the DRM of human podocytes overexpressing G1/G2-Vs. These findings suggest that altered sphingolipidsprofiles may contribute to the deranged functionality of the plasma membrane in APOL1 risk milieu.
Collapse
Affiliation(s)
- Manuela Valsecchi
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| | - Valentina Cazzetta
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy.,Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center - IRCCS, Rozzano, MI, Italy
| | - Ferdinando Oriolo
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center - IRCCS, Rozzano, MI, Italy
| | - Xiqian Lan
- Key Laboratory for Aging and Regenerative Medicine, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Rocco Piazza
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Moin A Saleem
- Pediatric Academic Renal Unit, University of Bristol, Bristol, UK
| | - Pravin C Singhal
- Institute of Molecular Medicine, Feinstein Institute for Medical Research and Zucker School of Medicine at Hofstra-Northwell, Hempstead, NY, USA
| | - Domenico Mavilio
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy.,Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center - IRCCS, Rozzano, MI, Italy
| | - Joanna Mikulak
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy.,Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center - IRCCS, Rozzano, MI, Italy
| | - Massimo Aureli
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy.
| |
Collapse
|
41
|
Scales SJ, Gupta N, De Mazière AM, Posthuma G, Chiu CP, Pierce AA, Hötzel K, Tao J, Foreman O, Koukos G, Oltrabella F, Klumperman J, Lin W, Peterson AS. Apolipoprotein L1-Specific Antibodies Detect Endogenous APOL1 inside the Endoplasmic Reticulum and on the Plasma Membrane of Podocytes. J Am Soc Nephrol 2020; 31:2044-2064. [PMID: 32764142 DOI: 10.1681/asn.2019080829] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 05/10/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND APOL1 is found in human kidney podocytes and endothelia. Variants G1 and G2 of the APOL1 gene account for the high frequency of nondiabetic CKD among African Americans. Proposed mechanisms of kidney podocyte cytotoxicity resulting from APOL1 variant overexpression implicate different subcellular compartments. It is unclear where endogenous podocyte APOL1 resides, because previous immunolocalization studies utilized overexpressed protein or commercially available antibodies that crossreact with APOL2. This study describes and distinguishes the locations of both APOLs. METHODS Immunohistochemistry, confocal and immunoelectron microscopy, and podocyte fractionation localized endogenous and transfected APOL1 using a large panel of novel APOL1-specific mouse and rabbit monoclonal antibodies. RESULTS Both endogenous podocyte and transfected APOL1 isoforms vA and vB1 (and a little of isoform vC) localize to the luminal face of the endoplasmic reticulum (ER) and to the cell surface, but not to mitochondria, endosomes, or lipid droplets. In contrast, APOL2, isoform vB3, and most vC of APOL1 localize to the cytoplasmic face of the ER and are consequently absent from the cell surface. APOL1 knockout podocytes do not stain for APOL1, attesting to the APOL1-specificity of the antibodies. Stable re-transfection of knockout podocytes with inducible APOL1-G0, -G1, and -G2 showed no differences in localization among variants. CONCLUSIONS APOL1 is found in the ER and plasma membrane, consistent with either the ER stress or surface cation channel models of APOL1-mediated cytotoxicity. The surface localization of APOL1 variants potentially opens new therapeutic targeting avenues.
Collapse
Affiliation(s)
- Suzie J Scales
- Department of Molecular Biology, Genentech, South San Francisco, California .,Department of Immunology, Genentech, South San Francisco, California
| | - Nidhi Gupta
- Department of Molecular Biology, Genentech, South San Francisco, California.,Department of Immunology, Genentech, South San Francisco, California
| | - Ann M De Mazière
- Section of Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - George Posthuma
- Section of Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Cecilia P Chiu
- Department of Antibody Engineering, Genentech, South San Francisco, California
| | - Andrew A Pierce
- Department of Pathology, Genentech, South San Francisco, California
| | - Kathy Hötzel
- Department of Pathology, Genentech, South San Francisco, California
| | - Jianhua Tao
- Department of Pathology, Genentech, South San Francisco, California
| | - Oded Foreman
- Department of Pathology, Genentech, South San Francisco, California
| | - Georgios Koukos
- Department of Molecular Biology, Genentech, South San Francisco, California
| | | | - Judith Klumperman
- Section of Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - WeiYu Lin
- Department of Antibody Engineering, Genentech, South San Francisco, California
| | - Andrew S Peterson
- Department of Molecular Biology, Genentech, South San Francisco, California
| |
Collapse
|
42
|
Gupta N, Wang X, Wen X, Moran P, Paluch M, Hass PE, Heidersbach A, Haley B, Kirchhofer D, Brezski RJ, Peterson AS, Scales SJ. Domain-Specific Antibodies Reveal Differences in the Membrane Topologies of Apolipoprotein L1 in Serum and Podocytes. J Am Soc Nephrol 2020; 31:2065-2082. [PMID: 32764138 DOI: 10.1681/asn.2019080830] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 05/10/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Circulating APOL1 lyses trypanosomes, protecting against human sleeping sickness. Two common African gene variants of APOL1, G1 and G2, protect against infection by species of trypanosomes that resist wild-type APOL1. At the same time, the protection predisposes humans to CKD, an elegant example of balanced polymorphism. However, the exact mechanism of APOL1-mediated podocyte damage is not clear, including APOL1's subcellular localization, topology, and whether the damage is related to trypanolysis. METHODS APOL1 topology in serum (HDL particles) and in kidney podocytes was mapped with flow cytometry, immunoprecipitation, and trypanolysis assays that tracked 170 APOL1 domain-specific monoclonal antibodies. APOL1 knockout podocytes confirmed antibody specificity. RESULTS APOL1 localizes to the surface of podocytes, with most of the pore-forming domain (PFD) and C terminus of the Serum Resistance Associated-interacting domain (SRA-ID), but not the membrane-addressing domain (MAD), being exposed. In contrast, differential trypanolytic blocking activity reveals that the MAD is exposed in serum APOL1, with less of the PFD accessible. Low pH did not detectably alter the gross topology of APOL1, as determined by antibody accessibility, in serum or on podocytes. CONCLUSIONS Our antibodies highlighted different conformations of native APOL1 topology in serum (HDL particles) and at the podocyte surface. Our findings support the surface ion channel model for APOL1 risk variant-mediated podocyte injury, as well as providing domain accessibility information for designing APOL1-targeted therapeutics.
Collapse
Affiliation(s)
- Nidhi Gupta
- Department of Molecular Biology, Genentech, South San Francisco, California.,Department of Immunology, Genentech, South San Francisco, California
| | - Xinhua Wang
- Department of Antibody Engineering, Genentech, South San Francisco, California
| | - Xiaohui Wen
- Department of Molecular Biology, Genentech, South San Francisco, California
| | - Paul Moran
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California
| | - Maciej Paluch
- Department of Protein Chemistry, Genentech, South San Francisco, California
| | - Philip E Hass
- Department of Protein Chemistry, Genentech, South San Francisco, California
| | - Amy Heidersbach
- Department of Molecular Biology, Genentech, South San Francisco, California
| | - Benjamin Haley
- Department of Molecular Biology, Genentech, South San Francisco, California
| | - Daniel Kirchhofer
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California
| | - Randall J Brezski
- Department of Antibody Engineering, Genentech, South San Francisco, California
| | - Andrew S Peterson
- Department of Molecular Biology, Genentech, South San Francisco, California
| | - Suzie J Scales
- Department of Molecular Biology, Genentech, South San Francisco, California .,Department of Immunology, Genentech, South San Francisco, California
| |
Collapse
|
43
|
Plasma-Based Proteomics Profiling of Patients with Hyperthyroidism after Antithyroid Treatment. Molecules 2020; 25:molecules25122831. [PMID: 32575434 PMCID: PMC7356574 DOI: 10.3390/molecules25122831] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/14/2020] [Accepted: 06/17/2020] [Indexed: 12/22/2022] Open
Abstract
Thyroid hormones critically modulate body homeostasis and haemostasis by regulating energy and metabolism. Previous studies have focused on individual pathways or proteins that are affected by increases in thyroid hormone levels, while an overall plasma proteomic signature of this increased level is lacking. Herein, an integrated untargeted proteomic approach with network analysis was used to identify changes in circulating proteins in the plasma proteome between hyperthyroid and euthyroid states. Plasma from 10 age-matched subjects at baseline (hyperthyroid) and post treatment with carbimazole (euthyroid) was compared by difference gel electrophoresis (DIGE) and matrix-assisted laser desorption/ionization time of flight (MALDI TOF) mass spectrometry (MS). A total of 20 proteins were identified with significant difference in abundance (analysis of variance (ANOVA) test, p ≤ 0.05; fold-change ≥ 1.5) between the two states (12 increased and 8 decreased in abundance in the hyperthyroid state). Twelve protein spots corresponding to ten unique proteins were significantly more abundant in the hyperthyroid state compared with the euthyroid state. These increased proteins were haptoglobin (HP), hemopexin (HPX), clusterin (CLU), apolipoprotein L1 (APOL1), alpha-1-B glycoprotein (A1BG), fibrinogen gamma chain (FGG), Ig alpha-1 chain C region (IGHA1), complement C6 (C6), leucine rich alpha 2 glycoprotein (LRG1), and carboxypeptidase N catalytic chain (CPN1). Eight protein spots corresponding to six unique proteins were significantly decreased in abundance in the hyperthyroid samples compared with euthyroid samples. These decreased proteins were apolipoprotein A1 (APOA1), inter-alpha-trypsin inhibitor heavy chain 4 (ITIH4), plasminogen (PLG), alpha-1 antitrypsin (SERPINA1), fibrinogen beta chain (FGB), and complement C1r subcomponent (C1R). The differentially abundant proteins were investigated by ingenuity pathway analysis (IPA). The network pathway identified related to infectious disease, inflammatory disease, organismal injury and abnormalities, and the connectivity map focused around two central nodes, namely the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and p38 mitogen-activated protein kinase (MAPK) pathways. The plasma proteome of patients with hyperthyroidism revealed differences in the abundance of proteins involved in acute phase response signaling, and development of a hypercoagulable and hypofibrinolytic state. Our findings enhance our existing knowledge of the altered proteins and associated biochemical pathways in hyperthyroidism.
Collapse
|
44
|
Wang K, Wu T, Chen Y, Song G, Chen Z. Prognostic Effect of Preoperative Apolipoprotein B Level in Surgical Patients with Clear Cell Renal Cell Carcinoma. Oncol Res Treat 2020; 43:340-345. [PMID: 32554963 DOI: 10.1159/000507964] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/17/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND The aim of this study was to assess the prognostic value of the preoperative apolipoprotein B (ApoB) level in surgical patients with clear cell renal cell carcinoma (ccRCC). MATERIALS AND METHODS The study included 307 ccRCC patients receiving radical or partial nephrectomy between 2003 and 2012 in our center. The correlations among the preoperative ApoB, clinicopathological parameters, and overall survival (OS) were evaluated. RESULTS A total of 193 males (62.9%) and 114 females (37.1%) with ccRCC who underwent radical or partial nephrectomy were enrolled in the present study. The OS at 5 years after the operation was 90.6% for all patients, 87.4% for the lower ApoB group, and 97.0% for the higher-ApoB group. The cause-specific survival (CSS) at 5 years after surgery was 90.2% for all patients, 86.7% for the lower-ApoB group, and 97.0% for the higher-ApoB group. A higher-ApoB level was related to a better OS and CSS in ccRCC patients (p = 0.001 and p < 0.001, respectively). In multivariate analysis, age >60 years (p = 0.008 and p = 0.023) and a lower Apo B level (p = 0.019 and p = 0.018) were independent prognostic factors for OS and CSS, respectively. CONCLUSIONS In the Apo apolipoprotein family, the preoperative ApoB level had an important clinical significance for predicting the prognosis and survival rate of ccRCC patients.
Collapse
Affiliation(s)
- Kun Wang
- Department of Surgical Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Tingchun Wu
- Department of Surgical Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yiming Chen
- Department of Surgical Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Guanglai Song
- Department of Surgical Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Zhen Chen
- Department of Surgical Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China,
| |
Collapse
|
45
|
Giovinazzo JA, Thomson RP, Khalizova N, Zager PJ, Malani N, Rodriguez-Boulan E, Raper J, Schreiner R. Apolipoprotein L-1 renal risk variants form active channels at the plasma membrane driving cytotoxicity. eLife 2020; 9:51185. [PMID: 32427098 PMCID: PMC7292663 DOI: 10.7554/elife.51185] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 05/14/2020] [Indexed: 12/25/2022] Open
Abstract
Recently evolved alleles of Apolipoprotein L-1 (APOL1) provide increased protection against African trypanosome parasites while also significantly increasing the risk of developing kidney disease in humans. APOL1 protects against trypanosome infections by forming ion channels within the parasite, causing lysis. While the correlation to kidney disease is robust, there is little consensus concerning the underlying disease mechanism. We show in human cells that the APOL1 renal risk variants have a population of active channels at the plasma membrane, which results in an influx of both Na+ and Ca2+. We propose a model wherein APOL1 channel activity is the upstream event causing cell death, and that the activate-state, plasma membrane-localized channel represents the ideal drug target to combat APOL1-mediated kidney disease.
Collapse
Affiliation(s)
- Joseph A Giovinazzo
- Department of Biological Sciences, Hunter College at City University of New York, New York, United States
| | - Russell P Thomson
- Department of Biological Sciences, Hunter College at City University of New York, New York, United States
| | - Nailya Khalizova
- Department of Biological Sciences, Hunter College at City University of New York, New York, United States
| | - Patrick J Zager
- Department of Ophthalmology, Margaret Dyson Vision Research Institute, Weill Cornell Medicine, New York, United States
| | | | - Enrique Rodriguez-Boulan
- Department of Ophthalmology, Margaret Dyson Vision Research Institute, Weill Cornell Medicine, New York, United States
| | - Jayne Raper
- Department of Biological Sciences, Hunter College at City University of New York, New York, United States
| | - Ryan Schreiner
- Department of Ophthalmology, Margaret Dyson Vision Research Institute, Weill Cornell Medicine, New York, United States
| |
Collapse
|
46
|
Abstract
Apolipoprotein L1 (APOL1) is a protein encoded by the APOL1 gene, found only in humans and several primates. Two variants encoding two different isoforms exist for APOL1, namely G1 and G2. These variants confer increased protection against trypanosome infection, and subsequent African sleeping sickness, and also increase the likelihood of renal disease in individuals of African ancestry. APOL1 mutations are associated with increased risk of chronic kidney disease, inflammation, and exacerbation of systemic lupus erythematosus-associated renal dysfunction. This review serves to outline the structure and function of APOL1, as well as its role in several disease outcomes.
Collapse
Affiliation(s)
- Shanel Raghubeer
- Biomedical Sciences, Cape Peninsula University of Technology-Bellville Campus, Cape Town, Western Cape, South Africa
| | - Tahir S Pillay
- Department of Chemical Pathology, University of Pretoria Faculty of Health Sciences, Pretoria, Gauteng, South Africa.,Division of Chemical Pathology, University of Cape Town, Rondebosch, Western Cape, South Africa
| | - Tandi Edith Matsha
- Biomedical Sciences, Cape Peninsula University of Technology-Bellville Campus, Cape Town, Western Cape, South Africa
| |
Collapse
|
47
|
Fang J, Yao X, Hou M, Duan M, Xing L, Huang J, Wang Y, Zhu B, Chen Q, Wang H. ApoL1 induces kidney inflammation through RIG-I/NF-κB activation. Biochem Biophys Res Commun 2020; 527:466-473. [PMID: 32336543 DOI: 10.1016/j.bbrc.2020.04.054] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 04/11/2020] [Indexed: 02/07/2023]
Abstract
The genetic variations of the apolipoprotein L1 (APOL1) gene are associated with non-diabetic kidney diseases. However, very little is known about the role of ApoL1 in glomerular damage. Here, we aimed to identify the function and mechanism of ApoL1 in glomerular damage. The mice were randomly divided into two groups: one group was intraperitoneally injected with phosphate buffer saline (PBS), while the other group was intraperitoneally injected with recombinant ApoL1 every other day for 3 months. Hematoxylin and eosin (HE) and periodic acid Schiff (PAS) staining were used to demonstrate the effects of ApoL1 on kidney inflammation and injury. Furthermore, quantitative real-time polymerase chain reaction (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA) analyses revealed that ApoL1-treated mice exhibited enhanced expression of various inflammation markers in the kidney and serum compared to the PBS-treated mice. Immunofluorescence staining revealed that ApoL1 accumulated in kidney podocytes. Treatment with ApoL1 dose-dependently increased the expression of inflammation markers and apoptotic markers. The abnormal gene expression associated with ApoL1-mediated podocyte inflammation was evaluated using microarray analysis. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses revealed that the upregulated genes were enriched in the inflammation-related processes, such as the RIG-I/NF-κB signaling pathway. Consistently, the knockdown of RIG-I significantly mitigated the ApoL1-induced upregulation of inflammatory and apoptotic markers in the human podocytes. Additionally, the ApoL1-induced glomerular damage was attenuated in AAV-shRIG-I mice. Therefore, the effects of ApoL1 on glomerular damage may be, at least partially, through inducing abnormal expression of inflammatory molecules, which may have important implications for treatment of kidney diseases.
Collapse
Affiliation(s)
- Ji Fang
- Department of Nephrology, Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, People's Republic of China
| | - Xingmei Yao
- Department of Nephrology, Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, People's Republic of China
| | - Mingqiang Hou
- Department of Urology, Xishui County People's Hospital, Guizhou Province, Guizhou, 564699, People's Republic of China
| | - Miao Duan
- Department of Urology, Xishui County People's Hospital, Guizhou Province, Guizhou, 564699, People's Republic of China
| | - Lina Xing
- Department of Nephrology, Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, People's Republic of China
| | - Jiebo Huang
- Department of Nephrology, Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, People's Republic of China
| | - Yunman Wang
- Department of Nephrology, Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, People's Republic of China
| | - Bingbing Zhu
- Department of Nephrology, Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, People's Republic of China
| | - Qiujing Chen
- Institute of Cardiovascular Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, People's Republic of China.
| | - Hao Wang
- Department of Nephrology, Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, People's Republic of China.
| |
Collapse
|
48
|
Uzureau S, Lecordier L, Uzureau P, Hennig D, Graversen JH, Homblé F, Mfutu PE, Oliveira Arcolino F, Ramos AR, La Rovere RM, Luyten T, Vermeersch M, Tebabi P, Dieu M, Cuypers B, Deborggraeve S, Rabant M, Legendre C, Moestrup SK, Levtchenko E, Bultynck G, Erneux C, Pérez-Morga D, Pays E. APOL1 C-Terminal Variants May Trigger Kidney Disease through Interference with APOL3 Control of Actomyosin. Cell Rep 2020; 30:3821-3836.e13. [PMID: 32187552 PMCID: PMC7090385 DOI: 10.1016/j.celrep.2020.02.064] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/17/2020] [Accepted: 02/14/2020] [Indexed: 11/18/2022] Open
Abstract
The C-terminal variants G1 and G2 of apolipoprotein L1 (APOL1) confer human resistance to the sleeping sickness parasite Trypanosoma rhodesiense, but they also increase the risk of kidney disease. APOL1 and APOL3 are death-promoting proteins that are partially associated with the endoplasmic reticulum and Golgi membranes. We report that in podocytes, either APOL1 C-terminal helix truncation (APOL1Δ) or APOL3 deletion (APOL3KO) induces similar actomyosin reorganization linked to the inhibition of phosphatidylinositol-4-phosphate [PI(4)P] synthesis by the Golgi PI(4)-kinase IIIB (PI4KB). Both APOL1 and APOL3 can form K+ channels, but only APOL3 exhibits Ca2+-dependent binding of high affinity to neuronal calcium sensor-1 (NCS-1), promoting NCS-1-PI4KB interaction and stimulating PI4KB activity. Alteration of the APOL1 C-terminal helix triggers APOL1 unfolding and increased binding to APOL3, affecting APOL3-NCS-1 interaction. Since the podocytes of G1 and G2 patients exhibit an APOL1Δ or APOL3KO-like phenotype, APOL1 C-terminal variants may induce kidney disease by preventing APOL3 from activating PI4KB, with consecutive actomyosin reorganization of podocytes.
Collapse
Affiliation(s)
- Sophie Uzureau
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Laurence Lecordier
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Pierrick Uzureau
- Laboratory of Experimental Medicine (ULB222), CHU Charleroi, Université Libre de Bruxelles, Montigny le Tilleul, Belgium
| | - Dorle Hennig
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Jonas H Graversen
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Fabrice Homblé
- Laboratory of Structure and Function of Biological Membranes, Université Libre de Bruxelles, 1050 Brussels, Belgium
| | - Pepe Ekulu Mfutu
- Pediatric Nephrology, University Hospital Leuven, 3000 Leuven, Belgium
| | | | - Ana Raquel Ramos
- Institute of Interdisciplinary Research in Human and Molecular Biology, Campus Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Rita M La Rovere
- Laboratory of Molecular and Cellular Signalling, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Tomas Luyten
- Laboratory of Molecular and Cellular Signalling, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Marjorie Vermeersch
- Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Patricia Tebabi
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Marc Dieu
- URBC-Narilis, University of Namur, 5000 Namur, Belgium
| | - Bart Cuypers
- Biomedical Sciences Department, Institute of Tropical Medicine, 2000 Antwerpen, Belgium; Adrem Data Lab, Department of Mathematics and Computer Science, University of Antwerp, 2000 Antwerpen, Belgium
| | - Stijn Deborggraeve
- Biomedical Sciences Department, Institute of Tropical Medicine, 2000 Antwerpen, Belgium
| | - Marion Rabant
- Adult Nephrology-Transplantation Department, Paris Hospitals and Paris Descartes University, 75006 Paris, France
| | - Christophe Legendre
- Pathology Department, Paris Hospitals and Paris Descartes University, 75006 Paris, France
| | - Søren K Moestrup
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense C, Denmark; Department of Biomedicine, University of Aarhus, 8000 Aarhus, Denmark
| | - Elena Levtchenko
- Pediatric Nephrology, University Hospital Leuven, 3000 Leuven, Belgium
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signalling, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Christophe Erneux
- Institute of Interdisciplinary Research in Human and Molecular Biology, Campus Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - David Pérez-Morga
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium; Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Etienne Pays
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium.
| |
Collapse
|
49
|
Chaudhary NS, Moore JX, Zakai NA, Judd SE, Naik RP, Limou S, Cushman M, Lange LA, Wang HE, Winkler CA, Irvin MR, Kopp JB, Gutiérrez OM. APOL1 Nephropathy Risk Alleles and Risk of Sepsis in Blacks. Clin J Am Soc Nephrol 2019; 14:1733-1740. [PMID: 31704668 PMCID: PMC6895473 DOI: 10.2215/cjn.04490419] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 09/30/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND OBJECTIVES apo L1 (APOL1) nephropathy risk alleles are associated with CKD in blacks. Although APOL1 has innate immune functions, little is known about the association of APOL1 genotypes with risk of infectious outcomes, such as sepsis. The objective of this study was to examine the associations of APOL1 nephropathy risk alleles with risk of sepsis in black adults. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We assessed the association of APOL1 risk alleles with incident sepsis in 10,366 black participants of the Reasons for Geographic and Racial Differences in Stroke study enrolled between 2003 and 2007 with follow-up through December 31, 2012. In Cox models adjusted for demographics, comorbid conditions, and principal components ancestry, we examined the association of APOL1 risk alleles with incident sepsis using recessive (comparing zero or one versus two risk alleles), dominant (zero versus one or two risk alleles), and additive genetic models. We also examined models stratified by diabetes and CKD status. RESULTS A total of 1320 (13%) participants had two APOL1 risk alleles, 4719 (46%) had one risk allele, and 4327 (42%) participants had zero risk alleles. A total of 306 sepsis events occurred over a median 6.5 years (interquartile range, 4.5-8.1). There were no statistically significant associations of APOL1 genotype with sepsis risk under recessive genetic models. APOL1 genotypes were associated with sepsis risk under dominant (hazard ratio, 1.55; 95% confidence interval, 1.13 to 2.11) and additive (hazard ratio per variant allele copy, 1.25; 95% confidence interval, 1.02 to 1.53) genetic models adjusted for covariates and ancestry. These associations did not vary by diabetes or CKD status (Pinteraction>0.10 for both). CONCLUSIONS In community-dwelling black adults, carriage of APOL1 nephropathy risk alleles are common and associated with higher risk of sepsis.
Collapse
Affiliation(s)
| | - Justin X. Moore
- Departments of Epidemiology
- Division of Public Health Sciences, Department of Surgery, Washington University, St. Louis, Missouri
| | - Neil A. Zakai
- Departments of Medicine and Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | | | - Rakhi P. Naik
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Sophie Limou
- Nantes University, French National Institute of Health and Medical Research, Center for Research in Transplantation and Immunology, Nantes, France
| | - Mary Cushman
- Departments of Medicine and Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Leslie A. Lange
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Henry E. Wang
- Department of Emergency Medicine, University of Texas Health Science Center at Houston, Houston, Texas
| | - Cheryl A. Winkler
- Basic Research Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland; and
| | | | - Jeffrey B. Kopp
- Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Orlando M. Gutiérrez
- Departments of Epidemiology
- Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
50
|
Abstract
Genetic variants in the APOL1 gene, found only in individuals of recent African ancestry, greatly increase risk of multiple types of kidney disease. These APOL1 kidney risk alleles are a rare example of genetic variants that are common but also have a powerful effect on disease susceptibility. These alleles rose to high frequency in sub-Saharan Africa because they conferred protection against pathogenic trypanosomes that cause African sleeping sickness. We consider the genetic evidence supporting the association between APOL1 and kidney disease across the range of clinical phenotypes in the APOL1 nephropathy spectrum. We then explore the origins of the APOL1 risk variants and evolutionary struggle between humans and trypanosomes at both the molecular and population genetic level. Finally, we survey the rapidly growing literature investigating APOL1 biology as elucidated from experiments in cell-based systems, cell-free systems, mouse and lower organism models of disease, and through illuminating natural experiments in humans.
Collapse
Affiliation(s)
- David J Friedman
- Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA; ,
| | - Martin R Pollak
- Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA; ,
| |
Collapse
|