1
|
Ytterbrink C, Shubbar E, Parris TZ, Langen B, Druid M, Schüler E, Strand SE, Åkerström B, Gram M, Helou K, Forssell-Aronsson E. Effects of Recombinant α 1-Microglobulin on Early Proteomic Response in Risk Organs after Exposure to 177Lu-Octreotate. Int J Mol Sci 2024; 25:7480. [PMID: 39000587 PMCID: PMC11242497 DOI: 10.3390/ijms25137480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
Recombinant α1-microglobulin (A1M) is proposed as a protector during 177Lu-octreotate treatment of neuroendocrine tumors, which is currently limited by bone marrow and renal toxicity. Co-administration of 177Lu-octreotate and A1M could result in a more effective treatment by protecting healthy tissue, but the radioprotective action of A1M is not fully understood. The aim of this study was to examine the proteomic response of kidneys and bone marrow early after 177Lu-octreotate and/or A1M administration. Mice were injected with 177Lu-octreotate and/or A1M, while control mice received saline or A1M vehicle solution. Bone marrow, kidney medulla, and kidney cortex were sampled after 24 h or 7 d. The differential protein expression was analyzed with tandem mass spectrometry. The dosimetric estimation was based on 177Lu activity in the kidney. PHLDA3 was the most prominent radiation-responsive protein in kidney tissue. In general, no statistically significant difference in the expression of radiation-related proteins was observed between the irradiated groups. Most canonical pathways were identified in bone marrow from the 177Lu-octreotate+A1M group. Altogether, a tissue-dependent proteomic response followed exposure to 177Lu-octreotate alone or together with A1M. Combining 177Lu-octreotate with A1M did not inhibit the radiation-induced protein expression early after exposure, and late effects should be further studied.
Collapse
Affiliation(s)
- Charlotte Ytterbrink
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden; (C.Y.); (E.S.); (M.D.)
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (T.Z.P.); (K.H.)
| | - Emman Shubbar
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden; (C.Y.); (E.S.); (M.D.)
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (T.Z.P.); (K.H.)
| | - Toshima Z. Parris
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (T.Z.P.); (K.H.)
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Britta Langen
- Section of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Malin Druid
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden; (C.Y.); (E.S.); (M.D.)
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (T.Z.P.); (K.H.)
| | - Emil Schüler
- Department of Radiation Physics, Division of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA;
| | - Sven-Erik Strand
- Department of Clinical Sciences Lund, Oncology, Lund University, 221 00 Lund, Sweden;
| | - Bo Åkerström
- Department of Clinical Sciences Lund, Infection Medicine, Lund University, 221 00 Lund, Sweden;
| | - Magnus Gram
- Department of Clinical Sciences Lund, Pediatrics, Lund University, 221 00 Lund, Sweden;
- Department of Neonatology, Skåne University Hospital, 222 42 Lund, Sweden
- Biofilms—Research Center for Biointerfaces, Department of Biomedical Science, Faculty of Health and Society, Malmö University, 205 06 Malmö, Sweden
| | - Khalil Helou
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (T.Z.P.); (K.H.)
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Eva Forssell-Aronsson
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden; (C.Y.); (E.S.); (M.D.)
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (T.Z.P.); (K.H.)
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| |
Collapse
|
2
|
Vanarsa K, Zhang T, Hutcheson J, Kumar SR, Nukala S, Inthavong H, Stanley B, Wu T, Mok CC, Saxena R, Mohan C. iTRAQ-based mass spectrometry screen to identify serum biomarkers in systemic lupus erythematosus. Lupus Sci Med 2024; 11:e000673. [PMID: 38782493 PMCID: PMC11116855 DOI: 10.1136/lupus-2022-000673] [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: 02/02/2022] [Accepted: 09/15/2022] [Indexed: 05/25/2024]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a complex systemic autoimmune disorder with no reliable serum biomarkers currently available other than autoantibodies. METHODS In the present study, isobaric tags for relative and absolute quantitation-based mass spectrometry was used to screen the sera of patients with SLE to uncover potential disease biomarkers. RESULTS 85 common proteins were identified, with 16 being elevated (≥1.3) and 23 being decreased (≤0.7) in SLE. Of the 16 elevated proteins, serum alpha-1-microglobulin/bikunin precursor (AMBP), zinc alpha-2 glycoprotein (AZGP) and retinol-binding protein 4 (RBP4) were validated in independent cross-sectional cohorts (Cohort I, N=52; Cohort II, N=117) using an orthogonal platform, ELISA. Serum AMBP, AZGP and RBP4 were validated to be significantly elevated in both patients with inactive SLE and patients with active SLE compared with healthy controls (HCs) (p<0.05, fold change >2.5) in Cohort I. All three proteins exhibited good discriminatory power for distinguishing active SLE and inactive SLE (area under the curve=0.82-0.96), from HCs. Serum AMBP exhibited the largest fold change in active SLE (5.96) compared with HCs and correlated with renal disease activity. The elevation in serum AMBP was validated in a second cohort of patients with SLE of different ethnic origins, correlating with serum creatinine (r=0.60, p<0.001). CONCLUSION Since serum AMBP is validated to be elevated in SLE and correlated with renal disease, the clinical utility of this novel biomarker warrants further analysis in longitudinal cohorts of patients with lupus and lupus nephritis.
Collapse
Affiliation(s)
- Kamala Vanarsa
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Ting Zhang
- University of Houston, Houston, Texas, USA
- Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | | | - Sneha Ravi Kumar
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA
| | | | - Haleigh Inthavong
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA
| | | | - Tianfu Wu
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - C C Mok
- Medicine, Tuen Mun Hospital, Hong Kong
| | - Ramesh Saxena
- The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chandra Mohan
- Department Biomedical Engineering, University of Houston, Houston, Texas, USA
| |
Collapse
|
3
|
Chen Z, Wu J, Han J, Wang Y, Ni L. UCST-Type Soluble Immobilized Cellulase: A New Strategy for the Efficient Degradation and Improved Recycling Performance of Wastepaper Cellulose. Molecules 2024; 29:1039. [PMID: 38474553 DOI: 10.3390/molecules29051039] [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: 02/04/2024] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
This paper reports an innovative study that aims to address key issues in the efficient recycling of wastepaper cellulose. The research team utilized the temperature-responsive upper critical solution temperature (UCST) polymer P(NAGA-b-DMA) in combination with the LytA label's affinity for choline analogs. This innovative approach enabled them to successfully develop a novel soluble immobilized enzyme, P(NAGA-b-DMA)-cellulase. This new enzyme has proven highly effective, significantly enhancing the degradation of wastepaper cellulose while demonstrating exceptional stability. Compared with the traditional insoluble immobilized cellulase, the enzyme showed a significant improvement in the pH, temperature stability, recycling ability, and storage stability. A kinetic parameter calculation showed that the enzymatic effectiveness of the soluble immobilized enzyme was much better than that of the traditional insoluble immobilized cellulase. After the immobilization reaction, the Michaelis constant of the immobilized enzyme was only increased by 11.5%. In the actual wastepaper degradation experiment, the immobilized enzyme was effectively used, and it was found that the degradation efficiency of wastepaper cellulose reached 80% of that observed in laboratory conditions. This novel, thermosensitive soluble immobilized cellulase can efficiently catalyze the conversion of wastepaper cellulose into glucose under suitable conditions, so as to further ferment into environmentally friendly biofuel ethanol, which provides a solution to solve the shortage of raw materials and environmental protection problems in the paper products industry.
Collapse
Affiliation(s)
- Zhaohui Chen
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Jiacong Wu
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Juan Han
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Yun Wang
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Liang Ni
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
4
|
Nourie N, Ghaleb R, Lefaucheur C, Louis K. Toward Precision Medicine: Exploring the Landscape of Biomarkers in Acute Kidney Injury. Biomolecules 2024; 14:82. [PMID: 38254682 PMCID: PMC10813773 DOI: 10.3390/biom14010082] [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: 12/16/2023] [Revised: 01/02/2024] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
Acute kidney injury (AKI) remains a complex challenge with diverse underlying pathological mechanisms and etiologies. Current detection methods predominantly rely on serum creatinine, which exhibits substantial limitations in specificity and poses the issue of late-stage detection of kidney injury. In this review, we propose an up-to-date and comprehensive summary of advancements that identified novel biomarker candidates in blood and urine and ideal criteria for AKI biomarkers such as renal injury specificity, mechanistic insight, prognostic capacity, and affordability. Recently identified biomarkers not only indicate injury location but also offer valuable insights into a range of pathological processes, encompassing reduced glomerular filtration rate, tubular function, inflammation, and adaptive response to injury. The clinical applications of AKI biomarkers are becoming extensive and serving as relevant tools in distinguishing acute tubular necrosis from other acute renal conditions. Also, these biomarkers can offer significant insights into the risk of progression to chronic kidney disease CKD and in the context of kidney transplantation. Integration of these biomarkers into clinical practice has the potential to improve early diagnosis of AKI and revolutionize the design of clinical trials, offering valuable endpoints for therapeutic interventions and enhancing patient care and outcomes.
Collapse
Affiliation(s)
- Nicole Nourie
- Department of Nephrology and Kidney Transplantation, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris, 75010 Paris, France
- Human Immunology and Immunopathology, Inserm UMR 976, Université Paris Cité, 75010 Paris, France
| | - Rita Ghaleb
- Faculty of Medicine, Saint Joseph University, Beirut 1104 2020, Lebanon
| | - Carmen Lefaucheur
- Department of Nephrology and Kidney Transplantation, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris, 75010 Paris, France
- Human Immunology and Immunopathology, Inserm UMR 976, Université Paris Cité, 75010 Paris, France
| | - Kevin Louis
- Department of Nephrology and Kidney Transplantation, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris, 75010 Paris, France
- Human Immunology and Immunopathology, Inserm UMR 976, Université Paris Cité, 75010 Paris, France
| |
Collapse
|
5
|
Kobayashi Y, Eguchi A, Imami K, Tempaku M, Izuoka K, Takase T, Kainuma K, Nagao M, Furuta N, Iwasa M, Nakagawa H, Fujisawa T, Togashi K. Circulating extracellular vesicles are associated with pathophysiological condition including metabolic syndrome-related dysmetabolism in children and adolescents with obesity. J Mol Med (Berl) 2024; 102:23-38. [PMID: 37874387 DOI: 10.1007/s00109-023-02386-5] [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: 03/20/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/25/2023]
Abstract
Obesity of children and adolescents (OCA) is often accompanied by metabolic syndrome (MetS), which often leads to adult obesity and subsequent complications, yet the entire pathophysiological response is not fully understood. The number and composition of circulating extracellular vesicles (EV) reflect overall patient condition; therefore, we investigated the pathophysiological condition of OCA, including MetS-associated dysmetabolism, using circulating EVs. In total, 107 children and adolescents with or without obesity (boys, n = 69; girls, n = 38; median age, 10 years) were enrolled. Circulating EV number and EV protein composition were assessed via flow cytometry and liquid chromatography tandem-mass spectrometry, respectively. In a multivariate analysis, relative body weight (standardized partial regression coefficient (SPRC) 0.469, P = 0.012) and serum triglyceride level (SPRC 0.548, P < 0.001) were detected as independent parameters correlating with circulating EV number. Proteomic analysis identified 31 upregulated and 45 downregulated EV proteins in OCA. Gene ontology analysis revealed upregulated proteins to be involved in various biological processes, including intracellular protein transport, protein folding, stress response, leukocyte activation, innate immune response, and platelet degranulation, which can modulate lipid and glucose metabolism, skeletal and cardiac muscle development, inflammation, immune response, carcinogenesis, and cancer progression. Notably, several identified EV proteins are involved in neuro-development, neurotransmitter release, and neuro-protective agents in OCA. Circulating EVs were derived from adipocytes, hepatocytes, B cell lymphocytes, and neurons. Circulating EV number is significantly associated with MetS-related dysmetabolism and the EV protein cargo carries a special "signature" that reflects the alteration of various biological processes under the pathophysiological condition of OCA. KEY MESSAGES: Circulating EV number correlates with physical and laboratory parameters for obesity in children and adolescents. Relative body weight and triglyceride are independent factors for increased circulating EVs. EV composition is significantly changed in obesity of children and adolescents. Identified EV composition changes associated with obesity and involves in metabolism, immune response, and cancer progression. Circulating EVs are partially derived from adipocyte, hepatocytes, B cells, and neurons.
Collapse
Affiliation(s)
- Yoshinao Kobayashi
- Center for Physical and Mental Health, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Akiko Eguchi
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan.
- JST, PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan.
- Biobank Center, Mie University Hospital, Tsu, Mie, 514-8507, Japan.
| | - Koshi Imami
- JST, PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Mina Tempaku
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Kiyora Izuoka
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Takafumi Takase
- Department of Pediatrics, National Hospital Organization Mie National Hospital, Tsu, Mie, 514-0125, Japan
| | - Keigo Kainuma
- Department of Pediatrics, National Hospital Organization Mie National Hospital, Tsu, Mie, 514-0125, Japan
| | - Mizuho Nagao
- Department of Pediatrics, National Hospital Organization Mie National Hospital, Tsu, Mie, 514-0125, Japan
| | - Noriko Furuta
- Center for Physical and Mental Health, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan
| | - Motoh Iwasa
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Hayato Nakagawa
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Takao Fujisawa
- Department of Pediatrics, National Hospital Organization Mie National Hospital, Tsu, Mie, 514-0125, Japan
| | - Kenji Togashi
- Department of Health and Physical Education, Faculty of Education, Mie University, Tsu, Mie, 514-8507, Japan
| |
Collapse
|
6
|
Bao H, Cao J, Chen M, Chen M, Chen W, Chen X, Chen Y, Chen Y, Chen Y, Chen Z, Chhetri JK, Ding Y, Feng J, Guo J, Guo M, He C, Jia Y, Jiang H, Jing Y, Li D, Li J, Li J, Liang Q, Liang R, Liu F, Liu X, Liu Z, Luo OJ, Lv J, Ma J, Mao K, Nie J, Qiao X, Sun X, Tang X, Wang J, Wang Q, Wang S, Wang X, Wang Y, Wang Y, Wu R, Xia K, Xiao FH, Xu L, Xu Y, Yan H, Yang L, Yang R, Yang Y, Ying Y, Zhang L, Zhang W, Zhang W, Zhang X, Zhang Z, Zhou M, Zhou R, Zhu Q, Zhu Z, Cao F, Cao Z, Chan P, Chen C, Chen G, Chen HZ, Chen J, Ci W, Ding BS, Ding Q, Gao F, Han JDJ, Huang K, Ju Z, Kong QP, Li J, Li J, Li X, Liu B, Liu F, Liu L, Liu Q, Liu Q, Liu X, Liu Y, Luo X, Ma S, Ma X, Mao Z, Nie J, Peng Y, Qu J, Ren J, Ren R, Song M, Songyang Z, Sun YE, Sun Y, Tian M, Wang S, Wang S, Wang X, Wang X, Wang YJ, Wang Y, Wong CCL, Xiang AP, Xiao Y, Xie Z, Xu D, Ye J, Yue R, Zhang C, Zhang H, Zhang L, Zhang W, Zhang Y, Zhang YW, Zhang Z, Zhao T, Zhao Y, Zhu D, Zou W, Pei G, Liu GH. Biomarkers of aging. SCIENCE CHINA. LIFE SCIENCES 2023; 66:893-1066. [PMID: 37076725 PMCID: PMC10115486 DOI: 10.1007/s11427-023-2305-0] [Citation(s) in RCA: 99] [Impact Index Per Article: 99.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/27/2023] [Indexed: 04/21/2023]
Abstract
Aging biomarkers are a combination of biological parameters to (i) assess age-related changes, (ii) track the physiological aging process, and (iii) predict the transition into a pathological status. Although a broad spectrum of aging biomarkers has been developed, their potential uses and limitations remain poorly characterized. An immediate goal of biomarkers is to help us answer the following three fundamental questions in aging research: How old are we? Why do we get old? And how can we age slower? This review aims to address this need. Here, we summarize our current knowledge of biomarkers developed for cellular, organ, and organismal levels of aging, comprising six pillars: physiological characteristics, medical imaging, histological features, cellular alterations, molecular changes, and secretory factors. To fulfill all these requisites, we propose that aging biomarkers should qualify for being specific, systemic, and clinically relevant.
Collapse
Affiliation(s)
- Hainan Bao
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Jiani Cao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Mengting Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Min Chen
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Chen
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Xiao Chen
- Department of Nuclear Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Yanhao Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yutian Chen
- The Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhiyang Chen
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Ageing and Regenerative Medicine, Jinan University, Guangzhou, 510632, China
| | - Jagadish K Chhetri
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yingjie Ding
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Junlin Feng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jun Guo
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Mengmeng Guo
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Chuting He
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Yujuan Jia
- Department of Neurology, First Affiliated Hospital, Shanxi Medical University, Taiyuan, 030001, China
| | - Haiping Jiang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Ying Jing
- Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Dingfeng Li
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China
| | - Jiaming Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jingyi Li
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Qinhao Liang
- College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China
| | - Rui Liang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300384, China
| | - Feng Liu
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, 510275, China
| | - Xiaoqian Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Zuojun Liu
- School of Life Sciences, Hainan University, Haikou, 570228, China
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Jianwei Lv
- School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Jingyi Ma
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Kehang Mao
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, 100871, China
| | - Jiawei Nie
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine (Shanghai), International Center for Aging and Cancer, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xinhua Qiao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xinpei Sun
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, 100101, China
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Jianfang Wang
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Qiaoran Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Siyuan Wang
- Clinical Research Institute, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Xuan Wang
- Hepatobiliary and Pancreatic Center, Medical Research Center, Beijing Tsinghua Changgung Hospital, Beijing, 102218, China
| | - Yaning Wang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuhan Wang
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Rimo Wu
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Kai Xia
- Center for Stem Cell Biologyand Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Fu-Hui Xiao
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yingying Xu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Haoteng Yan
- Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Liang Yang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
| | - Ruici Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yuanxin Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Yilin Ying
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- International Laboratory in Hematology and Cancer, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China
| | - Le Zhang
- Gerontology Center of Hubei Province, Wuhan, 430000, China
- Institute of Gerontology, Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weiwei Zhang
- Department of Cardiology, The Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, 100853, China
| | - Wenwan Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xing Zhang
- Key Laboratory of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhuo Zhang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
- Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Min Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Qingchen Zhu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zhengmao Zhu
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin, 300071, China
- Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Feng Cao
- Department of Cardiology, The Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, 100853, China.
| | - Zhongwei Cao
- State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Piu Chan
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Chang Chen
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Guobing Chen
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Guangzhou, 510000, China.
| | - Hou-Zao Chen
- Department of Biochemistryand Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China.
| | - Jun Chen
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191, China.
| | - Weimin Ci
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
| | - Bi-Sen Ding
- State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Qiurong Ding
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Feng Gao
- Key Laboratory of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, 100871, China.
| | - Kai Huang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Ageing and Regenerative Medicine, Jinan University, Guangzhou, 510632, China.
| | - Qing-Peng Kong
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China.
| | - Xin Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Baohua Liu
- School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen, 518060, China.
| | - Feng Liu
- Metabolic Syndrome Research Center, The Second Xiangya Hospital, Central South Unversity, Changsha, 410011, China.
| | - Lin Liu
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin, 300071, China.
- Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Institute of Translational Medicine, Tianjin Union Medical Center, Nankai University, Tianjin, 300000, China.
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China.
| | - Qiang Liu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China.
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Tianjin Institute of Immunology, Tianjin Medical University, Tianjin, 300070, China.
| | - Xingguo Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China.
| | - Yong Liu
- College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China.
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, China.
| | - Shuai Ma
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Xinran Ma
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Jing Nie
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yaojin Peng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Jie Ren
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Ruibao Ren
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine (Shanghai), International Center for Aging and Cancer, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- International Center for Aging and Cancer, Hainan Medical University, Haikou, 571199, China.
| | - Moshi Song
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Zhou Songyang
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, 510275, China.
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| | - Yi Eve Sun
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Yu Sun
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
- Department of Medicine and VAPSHCS, University of Washington, Seattle, WA, 98195, USA.
| | - Mei Tian
- Human Phenome Institute, Fudan University, Shanghai, 201203, China.
| | - Shusen Wang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300384, China.
| | - Si Wang
- Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| | - Xia Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| | - Xiaoning Wang
- Institute of Geriatrics, The second Medical Center, Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Yan-Jiang Wang
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| | - Yunfang Wang
- Hepatobiliary and Pancreatic Center, Medical Research Center, Beijing Tsinghua Changgung Hospital, Beijing, 102218, China.
| | - Catherine C L Wong
- Clinical Research Institute, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China.
| | - Andy Peng Xiang
- Center for Stem Cell Biologyand Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China.
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Yichuan Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Zhengwei Xie
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, 100101, China.
- Beijing & Qingdao Langu Pharmaceutical R&D Platform, Beijing Gigaceuticals Tech. Co. Ltd., Beijing, 100101, China.
| | - Daichao Xu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China.
| | - Jing Ye
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- International Laboratory in Hematology and Cancer, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China.
| | - Rui Yue
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Cuntai Zhang
- Gerontology Center of Hubei Province, Wuhan, 430000, China.
- Institute of Gerontology, Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Hongbo Zhang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Liang Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Yong Zhang
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China.
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China.
| | - Zhuohua Zhang
- Key Laboratory of Molecular Precision Medicine of Hunan Province and Center for Medical Genetics, Institute of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China.
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Tongbiao Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Yuzheng Zhao
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
- Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Dahai Zhu
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China.
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Gang Pei
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Biomedicine, The Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, 200070, China.
| | - Guang-Hui Liu
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
7
|
Min J, Foo ACY, Gabel SA, Perera L, DeRose EF, Pomés A, Pedersen LC, Mueller GA. Structural and ligand binding analysis of the pet allergens Can f 1 and Fel d 7. FRONTIERS IN ALLERGY 2023; 4:1133412. [PMID: 36960093 PMCID: PMC10028261 DOI: 10.3389/falgy.2023.1133412] [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: 12/28/2022] [Accepted: 02/16/2023] [Indexed: 03/09/2023] Open
Abstract
Introduction Pet lipocalins are respiratory allergens with a central hydrophobic ligand-binding cavity called a calyx. Molecules carried in the calyx by allergens are suggested to influence allergenicity, but little is known about the native ligands. Methods To provide more information on prospective ligands, we report crystal structures, NMR, molecular dynamics, and florescence studies of a dog lipocalin allergen Can f 1 and its closely related (and cross-reactive) cat allergen Fel d 7. Results Structural comparisons with reported lipocalins revealed that Can f 1 and Fel d 7 calyxes are open and positively charged while other dog lipocalin allergens are closed and negatively charged. We screened fatty acids as surrogate ligands, and found that Can f 1 and Fel d 7 bind multiple ligands with preferences for palmitic acid (16:0) among saturated fatty acids and oleic acid (18:1 cis-9) among unsaturated ones. NMR analysis of methyl probes reveals that conformational changes occur upon binding of pinolenic acid inside the calyx. Molecular dynamics simulation shows that the carboxylic group of fatty acids shuttles between two positively charged amino acids inside the Can f 1 and Fel d 7 calyx. Consistent with simulations, the stoichiometry of oleic acid-binding is 2:1 (fatty acid: protein) for Can f 1 and Fel d 7. Discussion The results provide valuable insights into the determinants of selectivity and candidate ligands for pet lipocalin allergens Can f 1 and Fel d 7.
Collapse
Affiliation(s)
- Jungki Min
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Alexander C. Y. Foo
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Scott A. Gabel
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Lalith Perera
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Eugene F. DeRose
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Anna Pomés
- Basic Research, InBio, Charlottesville, VA, United States
| | - Lars C. Pedersen
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Geoffrey A. Mueller
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC, United States
| |
Collapse
|
8
|
Berezin AA, Obradovic Z, Berezina TA, Boxhammer E, Lichtenauer M, Berezin AE. Cardiac Hepatopathy: New Perspectives on Old Problems through a Prism of Endogenous Metabolic Regulations by Hepatokines. Antioxidants (Basel) 2023; 12:antiox12020516. [PMID: 36830074 PMCID: PMC9951884 DOI: 10.3390/antiox12020516] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/12/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Cardiac hepatopathy refers to acute or chronic liver damage caused by cardiac dysfunction in the absence of any other possible causative reasons of liver injury. There is a large number of evidence of the fact that cardiac hepatopathy is associated with poor clinical outcomes in patients with acute or actually decompensated heart failure (HF). However, the currently dominated pathophysiological background does not explain a role of metabolic regulative proteins secreted by hepatocytes in progression of HF, including adverse cardiac remodeling, kidney injury, skeletal muscle dysfunction, osteopenia, sarcopenia and cardiac cachexia. The aim of this narrative review was to accumulate knowledge of hepatokines (adropin; fetuin-A, selenoprotein P, fibroblast growth factor-21, and alpha-1-microglobulin) as adaptive regulators of metabolic homeostasis in patients with HF. It is suggested that hepatokines play a crucial, causative role in inter-organ interactions and mediate tissue protective effects counteracting oxidative stress, inflammation, mitochondrial dysfunction, apoptosis and necrosis. The discriminative potencies of hepatokines for HF and damage of target organs in patients with known HF is under on-going scientific discussion and requires more investigations in the future.
Collapse
Affiliation(s)
- Alexander A. Berezin
- Internal Medicine Department, Zaporozhye Medical Academy of Postgraduate Education, 69000 Zaporozhye, Ukraine
- Klinik Barmelweid, Department of Psychosomatic Medicine and Psychotherapy, 5017 Barmelweid, Switzerland
| | - Zeljko Obradovic
- Klinik Barmelweid, Department of Psychosomatic Medicine and Psychotherapy, 5017 Barmelweid, Switzerland
| | - Tetiana A. Berezina
- Department of Internal Medicine & Nephrology, VitaCenter, 69000 Zaporozhye, Ukraine
| | - Elke Boxhammer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| | - Michael Lichtenauer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| | - Alexander E. Berezin
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
- Internal Medicine Department, Zaporozhye State Medical University, 69035 Zaporozhye, Ukraine
- Correspondence:
| |
Collapse
|
9
|
Zhang X, Lv J, Liu P, Xie X, Wang M, Liu D, Zhang H, Jin J. Poly-IgA Complexes and Disease Severity in IgA Nephropathy. Clin J Am Soc Nephrol 2021; 16:1652-1664. [PMID: 34607844 PMCID: PMC8729420 DOI: 10.2215/cjn.01300121] [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: 01/26/2021] [Accepted: 09/21/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND OBJECTIVES Poly-IgA immune complex formation and glomerular deposition play a key role in IgA nephropathy. Our study sought to develop a new methodology for one-step serologic detection of poly-IgA levels. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS A novel ELISA method using recombinant CD89 as a "capturing" probe was established for detecting poly-IgA immune complex in plasma. We applied semiquantitative measurements of these poly-IgA indices in patients recruited at Peking University First Hospital who had IgA nephropathy or other kidney disease types, as compared with healthy controls. The longitudinal trend of the poly-IgA index and the association with pathologic parameters and treatment responses were evaluated. Finally, we analyzed the molecular composition of poly-IgA complexes in patients by mass spectrometry. RESULTS Recombinant CD89-mounted ELISA plates specifically captured plasma poly-IgA. The levels of poly-IgA immune complex (26.7 [interquartile range (IQR) 17.1-42.6] U/ml) in IgA nephropathy were significantly higher than those in healthy controls (15.5 [IQR 10.7-20.0] U/ml; P<0.001) or in controls with non-IgA nephropathy disease (14.8 [IQR 10.5-21.9] U/ml; P<0.001). Higher levels of poly-IgA immune complex were associated with lower eGFR and worse kidney outcome. Accuracy parameters and concordant statistics showed good discrimination between IgA nephropathy and healthy controls based on poly-IgA index levels (area under the curve [AUC], 0.78; 95% confidence interval [95% CI], 0.72 to 0.83; P<0.001), significantly outperforming galactose-deficient IgA1 levels (AUC, 0.70; P=0.05). Corticosteroid and immunosuppressant treatments lowered poly-IgA indices. After a recombinant CD89-directed workflow in conjunction with mass spectrometry, we also analyzed the molecular composition of IgA immune complex in patients with IgA nephropathy. CONCLUSIONS Higher level of recombinant CD89-bound poly-IgA immune complex was associated with the severity of the disease and with treatment response to steroids and immunosuppressants.
Collapse
Affiliation(s)
- Xue Zhang
- Renal Division, Peking University First Hospital, Beijing, China,Peking University Institute of Nephrology, Beijing, China,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China,Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China,Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Jicheng Lv
- Renal Division, Peking University First Hospital, Beijing, China,Peking University Institute of Nephrology, Beijing, China,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China,Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China,Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Pan Liu
- Division of Nephrology and Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Xinfang Xie
- Division of Nephrology and Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Manliu Wang
- Renal Division, Peking University First Hospital, Beijing, China,Peking University Institute of Nephrology, Beijing, China,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China,Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China,Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Dan Liu
- Proteomics Laboratory, Medical and Health Analytical Center, Peking University, Beijing, China
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Beijing, China,Peking University Institute of Nephrology, Beijing, China,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China,Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China,Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Jin
- Division of Nephrology and Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
10
|
Ishiwata S, Matsue Y, Nakamura Y, Dotare T, Sunayama T, Suda S, Yatsu S, Kato T, Hiki M, Kasai T, Minamino T. Clinical and prognostic values of urinary alpha1-microglobulin as a tubular marker in acute heart failure. Int J Cardiol 2021; 338:115-120. [PMID: 34181994 DOI: 10.1016/j.ijcard.2021.06.041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 06/23/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Although urinary alpha-1-microglobulin has been used as a marker of tubular dysfunction, its clinical and prognostic values in patients with acute heart failure have not been validated. METHODS We analyzed 623 patients (74 ± 13 years old, 60.0% male) with acute heart failure in whom urinary alpha-1-microglobulin (A1MG) levels were measured as tubular markers at the time of admission. The primary endpoint was all-cause mortality. RESULTS The median levels of urinary alpha-1-microglobulin with and without correction for urinary creatinine concentration were 8.80 (interquartile range: 4.20-17.7) mg/dL and 12.9 (5.92-30.7) mg/gCr, respectively. Urinary A1MG levels were significantly correlated with all of beta-2-microglobulin (r = 0.77), N-acetyl-β-D-glucosaminidase (r = 0.51), and estimated glomerular filtration rate (r = -0.42); however, alpha-1-microglobulin was most often predicted using beta-2-microglobulin or N-acetyl-β-D-glucosaminidase. During the 488-day (interquartile range: 185-938 days) follow-up, 141 deaths occurred. Higher A1MG levels were associated with higher mortality even after adjustment for other covariates. Only A1MG, but not beta-2-microglobulin or N-acetyl-β-D-glucosaminidase, yielded incremental prognostic information in addition to the preexisting prognostic factors (net-reclassification improvement: 0.254, P = 0.023; integrated discrimination improvement: 0.015, P = 0.028). CONCLUSIONS In patients hospitalized due to acute heart failure, urinary alpha-1-microglobulin was a marker of tubular dysfunction. High alpha-1-microglobulin was associated with all-cause mortality independent of glomerular function and was a better predictor of mortality than urinary beta-2-microglobulin.
Collapse
Affiliation(s)
- Sayaki Ishiwata
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; Cardiovascular Respiratory Sleep Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Yuya Matsue
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; Cardiovascular Respiratory Sleep Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan.
| | - Yutaka Nakamura
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Taishi Dotare
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Tsutomu Sunayama
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Shoko Suda
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; Cardiovascular Respiratory Sleep Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Shoichiro Yatsu
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Takao Kato
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Masaru Hiki
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Takatoshi Kasai
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; Cardiovascular Respiratory Sleep Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; Sleep and Sleep-Disordered Breathing Center, Juntendo University Hospital, Tokyo 113-8421, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
11
|
Xu B, Zhu L, Wang Q, Zhao Y, Jia M, Shi S, Liu L, Lv J, Lai W, Ji J, Zhang H. Mass spectrometry-based screening identifies circulating immunoglobulinA-α1-microglobulin complex as potential biomarker in immunoglobulin A nephropathy. Nephrol Dial Transplant 2021; 36:782-792. [PMID: 33351144 DOI: 10.1093/ndt/gfaa352] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Immunoglobulin A nephropathy (IgAN) is characterized by predominant IgA deposition in the glomerular mesangium. Previous studies have proved that renal-deposited IgA in IgAN came from circulating IgA1-containing complexes (CICs). METHODS To explore the composition of CICs in IgAN, we isolated CICs from IgAN patients and healthy controls and then quantitatively analyzed them by mass spectrometry. Meanwhile, the isolated CICs were used to treat human mesangial cells to monitor mesangial cell injury. Using the protein content and injury effects, the key constituent in CICs was identified. Then the circulating levels of identified key constituent-IgA complex were detected in an independent population by an in-house-developed enzyme-linked immunosorbent assay. RESULTS By comparing the proteins of CICs between IgAN patients and controls, we found that 14 proteins showed significantly different levels. Among them, α1-microglobulin content in CICs was associated with not only in vitro mesangial cell proliferation and monocyte chemoattractant protein 1 secretion, but also in vivo estimated glomerular filtration rate (eGFR) levels and tubulointerstitial lesions in IgAN patients. Moreover, we found α1-microglobulin was prone to bind aberrant glycosylated IgA1. Additionally, elevated circulating IgA-α1-microglobulin complex levels were detected in an independent IgAN population and IgA-α1-microglobulin complex levels were correlated with hypertension, eGFR levels and Oxford T- scores in these IgAN patients. CONCLUSIONS Our results suggest that the IgA-α1-microglobulin complex is an important constituent in CICs and that circulating IgA-α1-microglobulin complex detection might serve as a potential noninvasive biomarker detection method for IgAN.
Collapse
Affiliation(s)
- Boyang Xu
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, China, and Peking University Institute of Nephrology, Beijing, China.,Key Laboratory of Renal Disease (Peking University), National Health Commission, Beijing, China.,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China.,Research Unit of Diagnosis and Treatment of Immune-Mediated Kidney Disease, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Zhu
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, China, and Peking University Institute of Nephrology, Beijing, China.,Key Laboratory of Renal Disease (Peking University), National Health Commission, Beijing, China.,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China.,Research Unit of Diagnosis and Treatment of Immune-Mediated Kidney Disease, Chinese Academy of Medical Sciences, Beijing, China
| | - Qingsong Wang
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, China
| | - Yanfeng Zhao
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, China, and Peking University Institute of Nephrology, Beijing, China.,Key Laboratory of Renal Disease (Peking University), National Health Commission, Beijing, China.,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China.,Research Unit of Diagnosis and Treatment of Immune-Mediated Kidney Disease, Chinese Academy of Medical Sciences, Beijing, China
| | - Meng Jia
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, China, and Peking University Institute of Nephrology, Beijing, China.,Key Laboratory of Renal Disease (Peking University), National Health Commission, Beijing, China.,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China.,Research Unit of Diagnosis and Treatment of Immune-Mediated Kidney Disease, Chinese Academy of Medical Sciences, Beijing, China
| | - Sufang Shi
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, China, and Peking University Institute of Nephrology, Beijing, China.,Key Laboratory of Renal Disease (Peking University), National Health Commission, Beijing, China.,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China.,Research Unit of Diagnosis and Treatment of Immune-Mediated Kidney Disease, Chinese Academy of Medical Sciences, Beijing, China
| | - Lijun Liu
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, China, and Peking University Institute of Nephrology, Beijing, China.,Key Laboratory of Renal Disease (Peking University), National Health Commission, Beijing, China.,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China.,Research Unit of Diagnosis and Treatment of Immune-Mediated Kidney Disease, Chinese Academy of Medical Sciences, Beijing, China
| | - Jicheng Lv
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, China, and Peking University Institute of Nephrology, Beijing, China.,Key Laboratory of Renal Disease (Peking University), National Health Commission, Beijing, China.,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China.,Research Unit of Diagnosis and Treatment of Immune-Mediated Kidney Disease, Chinese Academy of Medical Sciences, Beijing, China
| | - Wenjia Lai
- Chinese Academy of Sciences Key Laboratory of Standardization and Measurement for Nanotechnology, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Jianguo Ji
- State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, Beijing, China
| | - Hong Zhang
- Department of Medicine, Renal Division, Peking University First Hospital, Beijing, China, and Peking University Institute of Nephrology, Beijing, China.,Key Laboratory of Renal Disease (Peking University), National Health Commission, Beijing, China.,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China.,Research Unit of Diagnosis and Treatment of Immune-Mediated Kidney Disease, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
12
|
Bergwik J, Kristiansson A, Allhorn M, Gram M, Åkerström B. Structure, Functions, and Physiological Roles of the Lipocalin α 1-Microglobulin (A1M). Front Physiol 2021; 12:645650. [PMID: 33746781 PMCID: PMC7965949 DOI: 10.3389/fphys.2021.645650] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/12/2021] [Indexed: 12/30/2022] Open
Abstract
α1-microglobulin (A1M) is found in all vertebrates including humans. A1M was, together with retinol-binding protein and β-lactoglobulin, one of the three original lipocalins when the family first was proposed in 1985. A1M is described as an antioxidant and tissue cleaning protein with reductase, heme- and radical-binding activities. These biochemical properties are driven by a strongly electronegative surface-exposed thiol group, C34, on loop 1 of the open end of the lipocalin barrel. A1M has been shown to have protective effects in vitro and in vivo in cell-, organ-, and animal models of oxidative stress-related medical conditions. The gene coding for A1M is unique among lipocalins since it is flanked downstream by four exons coding for another non-lipocalin protein, bikunin, and is consequently named α1-microglobulin-bikunin precursor gene (AMBP). The precursor is cleaved in the Golgi, and A1M and bikunin are secreted from the cell separately. Recent publications have suggested novel physiological roles of A1M in regulation of endoplasmic reticulum activities and erythrocyte homeostasis. This review summarizes the present knowledge of the structure and functions of the lipocalin A1M and presents a current model of its biological role(s).
Collapse
Affiliation(s)
- Jesper Bergwik
- Department of Clinical Sciences, Section for Infection Medicine, Lund University, Lund, Sweden
| | - Amanda Kristiansson
- Department of Clinical Sciences, Section for Infection Medicine, Lund University, Lund, Sweden.,Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Maria Allhorn
- Department of Clinical Sciences, Section for Infection Medicine, Lund University, Lund, Sweden
| | - Magnus Gram
- Department of Clinical Sciences, Pediatrics, Lund University, Lund, Sweden
| | - Bo Åkerström
- Department of Clinical Sciences, Section for Infection Medicine, Lund University, Lund, Sweden
| |
Collapse
|
13
|
Gu Z, Huang Y, Yang F, Tang S, Sun J, Chen T, Liu F, Wu H, Wang J. The application of neutrophil gelatin-related lipid delivery protein in evaluation of renal function, nutrition, anemia and inflammation in patients with CKD. Nephrol Ther 2021; 17:35-41. [PMID: 33468440 DOI: 10.1016/j.nephro.2020.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 09/15/2020] [Accepted: 10/11/2020] [Indexed: 01/23/2023]
Abstract
OBJECTIVE To investigate the role of neutrophil gelatinase-associated lipocalin in the evaluation of renal function, nutrition, anemia and inflammation in patients with chronic kidney diseases. MATERIALS AND METHODS A total of 302 patients with chronic kidney diseases were selected, and their clinical data, blood neutrophil gelatinase-associated lipocalin levels, renal function, nutrition, anemia, inflammation and calcium, and phosphorus metabolism were analyzed. RESULT Serum neutrophil gelatinase-associated lipocalin level increased with the progression of chronic kidney diseases. Higher neutrophil gelatinase-associated lipocalin levels were observed in patients with chronic kidney diseases stage 3b compared with healthy individuals (P<0.05), while the patients with chronic kidney diseases stage 5 showed higher levels compared with other chronic kidney diseases stages (P<0.01). Moreover, the ROC curve showed that neutrophil gelatinase-associated lipocalin had a better diagnostic performance from the chronic kidney diseases stage 3b to 5 (P<0.05). In addition, the serum neutrophil gelatinase-associated lipocalin levels in patient with chronic kidney diseases were negatively correlated with body mass index, number of red blood cells, hemoglobin, transferrin, the estimatedglomerular filtration rate (eGFR), serum calcium (P<0.01); and were positively correlated with mean arterial blood pressure, blood BUN, SCr and alpha 1 microglobulin, beta 2 microglobulin, urinary inhibition C, homocysteine, PTH levels, neutrophils ratio, free serum ferritin and c-reactive protein (P<0.01); while no significant correlation was found with gender, and age (P>0.05). CONCLUSION Serum neutrophil gelatinase-associated lipocalin levels are closely related to renal function injury, inflammatory response and anemia-related indicators in patients with chronic kidney diseases, and thus could be used as a diagnostic biomarker for evaluating the degree of renal injury and related complications in patients with chronic kidney diseases.
Collapse
Affiliation(s)
- Zhoupeng Gu
- Department of Nephropathy, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China
| | - Yan Huang
- Department of Nephropathy, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China
| | - Fengjie Yang
- Department of Nephropathy, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China
| | - Shiqi Tang
- Department of Nephropathy, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China
| | - Juan Sun
- Department of Nephropathy, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China
| | - Tianru Chen
- Department of Nephropathy, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China
| | - Fang Liu
- Department of Nephropathy, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China
| | - Hong Wu
- Department of Nephropathy, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China; Department of anesthesiology, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China
| | - Jianwen Wang
- Department of Nephropathy, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China.
| |
Collapse
|
14
|
Chambers IG, Willoughby MM, Hamza I, Reddi AR. One ring to bring them all and in the darkness bind them: The trafficking of heme without deliverers. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2021; 1868:118881. [PMID: 33022276 PMCID: PMC7756907 DOI: 10.1016/j.bbamcr.2020.118881] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/22/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023]
Abstract
Heme, as a hydrophobic iron-containing organic ring, is lipid soluble and can interact with biological membranes. The very same properties of heme that nature exploits to support life also renders heme potentially cytotoxic. In order to utilize heme, while also mitigating its toxicity, cells are challenged to tightly control the concentration and bioavailability of heme. On the bright side, it is reasonable to envision that, analogous to other transition metals, a combination of membrane-bound transporters, soluble carriers, and chaperones coordinate heme trafficking to subcellular compartments. However, given the dual properties exhibited by heme as a transition metal and lipid, it is compelling to consider the dark side: the potential role of non-proteinaceous biomolecules including lipids and nucleic acids that bind, sequester, and control heme trafficking and bioavailability. The emergence of inter-organellar membrane contact sites, as well as intracellular vesicles derived from various organelles, have raised the prospect that heme can be trafficked through hydrophobic channels. In this review, we aim to focus on heme delivery without deliverers - an alternate paradigm for the regulation of heme homeostasis through chaperone-less pathways for heme trafficking.
Collapse
Affiliation(s)
- Ian G Chambers
- Department of Animal and Avian Sciences, Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20740, United States of America
| | - Mathilda M Willoughby
- School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, United States of America
| | - Iqbal Hamza
- Department of Animal and Avian Sciences, Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20740, United States of America.
| | - Amit R Reddi
- School of Chemistry and Biochemistry, Parker Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, United States of America.
| |
Collapse
|
15
|
Zhang C, Luo Y, Zhang Z, Zhang Z, Zhang G, Wang F, Che Y, Fang L, Zhang Y, Sun N, He J. Identification of a Prognostic Immune Signature for Esophageal Squamous Cell Carcinoma to Predict Survival and Inflammatory Landscapes. Front Cell Dev Biol 2020; 8:580005. [PMID: 33392181 PMCID: PMC7773787 DOI: 10.3389/fcell.2020.580005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 09/03/2020] [Indexed: 01/06/2023] Open
Abstract
Immunotherapy has achieved success in the treatment of esophageal squamous cell carcinoma (ESCC). However, studies concerning immune phenotypes within the ESCC microenvironment and their relationship with prognostic outcomes are limited. We constructed and validated an individual immune-related risk signature for patients with ESCC. We collected 196 ESCC cases, including 119 samples from our previous public data (GSE53624) to use as a training set and an independent cohort with 77 quantitative real-time polymerase chain reaction (qRT-PCR) data, which we used for validation. Head and neck squamous cell carcinoma (HNSCC) and lung squamous cell carcinoma (LUSC) cohorts were also collected for validation. A least absolute shrinkage and selection operator (LASSO) model and a stepwise Cox proportional hazards regression model were used to construct the immune-specific signature. The potential mechanism and inflammatory landscapes of the signature were explored using bioinformatics and immunofluorescence assay methods. This signature predicted different prognoses in clinical subgroups and the independent cohort, as well as in patients with HNSCC and LUSC. Further exploration revealed that the signature was associated with specific inflammatory activities (activation of macrophages and T-cell signaling transduction). Additionally, high-risk patients exhibited distinctive immune checkpoints panel and higher regulatory T cell and fibroblast infiltration. This signature served as an independent prognostic factor in ESCC. This was the first applicable immune-related risk signature for ESCC. Our results furnished new hints of immune profiling of ESCC, which may provide some clues to further optimize associated cancer immunotherapies.
Collapse
Affiliation(s)
- Chaoqi Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yuejun Luo
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhen Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhihui Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Guochao Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Feng Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yun Che
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Lingling Fang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
Youssef L, Erlandsson L, Åkerström B, Miranda J, Paules C, Crovetto F, Crispi F, Gratacos E, Hansson SR. Hemopexin and α1-microglobulin heme scavengers with differential involvement in preeclampsia and fetal growth restriction. PLoS One 2020; 15:e0239030. [PMID: 32915914 PMCID: PMC7485876 DOI: 10.1371/journal.pone.0239030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 08/30/2020] [Indexed: 12/22/2022] Open
Abstract
Hemopexin and α1-microglobulin act as scavengers to eliminate free heme-groups responsible for hemoglobin-induced oxidative stress. The present study evaluated maternal and fetal plasma concentrations of these scavengers in the different phenotypes of placenta-mediated disorders. Singleton pregnancies with normotensive fetal growth restriction [FGR] (n = 47), preeclampsia without FGR (n = 45) and preeclampsia with FGR (n = 51) were included prospectively as well as uncomplicated pregnancies (n = 49). Samples were collected at delivery and ELISA analysis was applied to measure the hemopexin and α1-microglobulin concentrations. In maternal blood in preeclampsia with and without FGR, hemopexin was significantly lower (p = 0.003 and p<0.001, respectively) and α1-microglobulin was significantly higher (p<0.001 in both) whereas no difference existed in normotensive FGR mothers compared to controls. In contrast, in fetal blood in growth restricted fetuses with and without preeclampsia, both hemopexin and α1-microglobulin were significantly lower (p<0.001 and p = 0.001 for hemopexin, p = 0.016 and p = 0.013 for α1-microglobulin, respectively) with no difference in fetuses from preeclampsia without FGR in comparison to controls. Thus, hemopexin and α1-microglobulin present significantly altered concentrations in maternal blood in the maternal disease -preeclampsia- and in cord blood in the fetal disease -FGR-, which supports their differential role in placenta-mediated disorders in accordance with the clinical presentation of these disorders.
Collapse
Affiliation(s)
- Lina Youssef
- Section of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- * E-mail: ,
| | - Lena Erlandsson
- Section of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Bo Åkerström
- Section of infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jezid Miranda
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Cristina Paules
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Francesca Crovetto
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain
| | - Fatima Crispi
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain
| | - Eduard Gratacos
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain
| | - Stefan R. Hansson
- Section of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Skåne University Hospital, Lund/Malmö, Sweden
| |
Collapse
|
17
|
Jotwani V, Garimella PS, Katz R, Malhotra R, Bates J, Cheung AK, Chonchol M, Drawz PE, Freedman BI, Haley WE, Killeen AA, Punzi H, Sarnak MJ, Segal MS, Shlipak MG, Ix JH. Tubular Biomarkers and Chronic Kidney Disease Progression in SPRINT Participants. Am J Nephrol 2020; 51:797-805. [PMID: 32906135 DOI: 10.1159/000509978] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/05/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Kidney tubular atrophy on biopsy is a strong predictor of chronic kidney disease (CKD) progression, but tubular health is poorly quantified by traditional measures including estimated glomerular filtration rate (eGFR) and albuminuria. We hypothesized that urinary biomarkers of impaired tubule function would be associated with faster eGFR declines in persons with CKD. METHODS We measured baseline urine concentrations of uromodulin, β2-microglobulin (β2m), and α1-microglobulin (α1m) among 2,428 participants of the Systolic Blood Pressure Intervention Trial with an eGFR <60 mL/min/1.73 m2. We used linear mixed models to evaluate biomarker associations with annualized relative change in eGFR, stratified by randomization arm. RESULTS At baseline, the mean age was 73 ± 9 years and eGFR was 46 ± 11 mL/min/1.73 m2. In the standard blood pressure treatment arm, each 2-fold higher urinary uromodulin was associated with slower % annual eGFR decline (0.34 [95% CI: 0.08, 0.60]), whereas higher urinary β2m was associated with faster % annual eGFR decline (-0.10 [95% CI: -0.18, -0.02]) in multivariable-adjusted models including baseline eGFR and albuminuria. Associations were weaker and did not reach statistical significance in the intensive blood pressure treatment arm for either uromodulin (0.11 [-0.13, 0.35], p value for interaction by treatment arm = 0.045) or β2m (-0.01 [-0.08, 0.08], p value for interaction = 0.001). Urinary α1m was not independently associated with eGFR decline in the standard (0.01 [-0.22, 0.23]) or intensive (0.03 [-0.20, 0.25]) arm. CONCLUSIONS Among trial participants with hypertension and CKD, baseline measures of tubular function were associated with subsequent declines in kidney function, although these associations were diminished by intensive blood pressure control.
Collapse
Affiliation(s)
- Vasantha Jotwani
- Department of Medicine, San Francisco VA Medical Health Care System, San Francisco, California, USA,
- Kidney Health Research Collaborative, San Francisco VA Medical Center and University of California, San Francisco, California, USA,
| | - Pranav S Garimella
- Department of Medicine, University of California, San Diego, California, USA
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Ronit Katz
- Kidney Research Institute, University of Washington, Seattle, Washington, USA
| | - Rakesh Malhotra
- Department of Medicine, University of California, San Diego, California, USA
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Jeffrey Bates
- Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
| | - Alfred K Cheung
- Department of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Michel Chonchol
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Paul E Drawz
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Barry I Freedman
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - William E Haley
- Department of Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Anthony A Killeen
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Henry Punzi
- Punzi Medical Center, Trinity Hypertension Research Institute, Carollton, Texas, USA
| | - Mark J Sarnak
- Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Mark S Segal
- Department of Medicine, University of Florida, Gainesville, Florida, USA
| | - Michael G Shlipak
- Department of Medicine, San Francisco VA Medical Health Care System, San Francisco, California, USA
- Kidney Health Research Collaborative, San Francisco VA Medical Center and University of California, San Francisco, California, USA
| | - Joachim H Ix
- Department of Medicine, University of California, San Diego, California, USA
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| |
Collapse
|
18
|
Carlsson MLR, Kristiansson A, Bergwik J, Kanagarajan S, Bülow L, Åkerström B, Zhu LH. Expression, Purification and Initial Characterization of Functional α 1-Microglobulin (A1M) in Nicotiana benthamiana. FRONTIERS IN PLANT SCIENCE 2020; 11:593773. [PMID: 33363557 PMCID: PMC7752767 DOI: 10.3389/fpls.2020.593773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/12/2020] [Indexed: 05/08/2023]
Abstract
α1-Microglobulin (A1M) is a small glycoprotein that belongs to the lipocalin protein family. A major biological role of A1M is to protect cells and tissues against oxidative damage by clearing free heme and reactive oxygen species. Because of this, the protein has attracted great interest as a potential pharmaceutical candidate for treatment of acute kidney injury and preeclampsia. The aim of this study was to explore the possibility of expressing human A1M in plants through transient gene expression, as an alternative or complement to other expression systems. E. coli, insect and mammalian cell culture have previously been used for recombinant A1M (rA1M) or A1M production, but these systems have various drawbacks, including additional complication and expense in refolding for E. coli, while insect produced rA1M is heavily modified with chromophores and mammalian cell culture has been used only in analytical scale. For that purpose, we have used a viral vector (pJL-TRBO) delivered by Agrobacterium for expression of three modified A1M gene variants in the leaves of N. benthamiana. The results showed that these modified rA1M protein variants, A1M-NB1, A1M-NB2 and A1M-NB3, targeted to the cytosol, ER and extracellular space, respectively, were successfully expressed in the leaves, which was confirmed by SDS-PAGE and Western blot analysis. The cytosol accumulated A1M-NB1 was selected for further analysis, as it appeared to have a higher yield than the other variants, and was purified with a yield of ca. 50 mg/kg leaf. The purified protein had the expected structural and functional properties, displaying heme-binding capacity and capacity of protecting red blood cells against stress-induced cell death. The protein also carried bound chromophores, a characteristic feature of A1M and an indicator of a capacity to bind small molecules. The study showed that expression of the functional protein in N. benthamiana may be an attractive alternative for production of rA1M for pharmaceutical purposes and a basis for future research on A1M structure and function.
Collapse
Affiliation(s)
- Magnus L. R. Carlsson
- Department of Plant Breeding, Swedish University of Agricultural Sciences, Alnarp, Sweden
| | - Amanda Kristiansson
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jesper Bergwik
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Selvaraju Kanagarajan
- Department of Plant Breeding, Swedish University of Agricultural Sciences, Alnarp, Sweden
| | - Leif Bülow
- Division of Pure and Applied Biochemistry, Department of Chemistry, Lund University, Lund, Sweden
| | - Bo Åkerström
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Li-Hua Zhu
- Department of Plant Breeding, Swedish University of Agricultural Sciences, Alnarp, Sweden
- *Correspondence: Li-Hua Zhu,
| |
Collapse
|
19
|
Garimella PS, Lee AK, Ambrosius WT, Bhatt U, Cheung AK, Chonchol M, Craven T, Hawfield AT, Jotwani V, Killeen A, Punzi H, Sarnak MJ, Wall BM, Ix JH, Shlipak MG. Markers of kidney tubule function and risk of cardiovascular disease events and mortality in the SPRINT trial. Eur Heart J 2019; 40:3486-3493. [PMID: 31257404 PMCID: PMC6837159 DOI: 10.1093/eurheartj/ehz392] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/11/2019] [Accepted: 05/21/2019] [Indexed: 12/31/2022] Open
Abstract
AIMS Biomarkers of kidney tubule injury, inflammation and fibrosis have been studied extensively and established as risk markers of adverse kidney and cardiovascular disease (CVD) outcomes. However, associations of markers of kidney tubular function with adverse clinical events have not been well studied, especially in persons with chronic kidney disease (CKD). METHODS AND RESULTS Using a sample of 2377 persons with CKD at the baseline Systolic Blood Pressure Intervention Trial (SPRINT) visit, we evaluated the association of three urine tubular function markers, alpha-1 microglobulin (α1m), beta-2 microglobulin (β2m), and uromodulin, with a composite CVD endpoint (myocardial infarction, acute coronary syndrome, stroke, acute decompensated heart failure, or death from cardiovascular causes) and mortality using Cox proportional hazards regression, adjusted for baseline estimated glomerular filtration rate (eGFR), albuminuria, and CVD risk factors. In unadjusted analysis, over a median follow-up of 3.8 years, α1m and β2m had positive associations with composite CVD events and mortality, whereas uromodulin had an inverse association with risk for both outcomes. In multivariable analysis including eGFR and albuminuria, a two-fold higher baseline concentration of α1m was associated with higher risk of CVD [hazard ratio (HR) 1.25; 95% confidence interval (CI): 1.10-1.45] and mortality (HR 1.25; 95% CI: 1.10-1.46), whereas β2m had no association with either outcome. A two-fold higher uromodulin concentration was associated with lower CVD risk (HR 0.79; 95% CI: 0.68-0.90) but not mortality (HR 0.86; 95% CI: 0.73-1.01) after adjusting for similar confounders. CONCLUSION Among non-diabetic persons with CKD, biomarkers of tubular function are associated with CVD events and mortality independent of glomerular function and albuminuria.
Collapse
Affiliation(s)
- Pranav S Garimella
- Division of Nephrology and Hypertension, Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Alexandra K Lee
- Division of General Internal Medicine, San Francisco VA Medical Center, San Francisco, CA, USA
| | - Walter T Ambrosius
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Udayan Bhatt
- Division of Nephrology, Ohio State University, Columbus, OH, USA
| | - Alfred K Cheung
- Division of Nephrology & Hypertension, Department of Internal Medicine, Medical Service, University of Utah, Veterans Affairs Salt Lake City Healthcare System, Salt Lake City, UT, USA
| | - Michel Chonchol
- Division of Nephrology & Hypertension, Department of Medicine, University of Colorado, Denver, CO, USA
| | - Timothy Craven
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Amret T Hawfield
- Department of Internal Medicine, Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Vasantha Jotwani
- Kidney Health Research Collaborative, San Francisco VA Medical Center, University of California, San Francisco, CA, USA
| | - Anthony Killeen
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | | | - Mark J Sarnak
- Division of Nephrology, Tufts Medical Center, Boston, MA, USA
| | - Barry M Wall
- Division of Nephrology, University of Tennessee, Memphis, TN, USA
| | - Joachim H Ix
- Division of Nephrology and Hypertension, Department of Medicine, University of California San Diego, San Diego, CA, USA
- Division of Preventive Medicine, Department of Family Medicine and Public Health, University of California San Diego, San Diego, CA, USA
- Nephrology Section, Veterans Affairs San Diego Healthcare System, La Jolla, CA, USA
| | - Michael G Shlipak
- Division of General Internal Medicine, San Francisco VA Medical Center, San Francisco, CA, USA
- Kidney Health Research Collaborative, San Francisco VA Medical Center, University of California, San Francisco, CA, USA
| |
Collapse
|
20
|
Lee AK, Katz R, Jotwani V, Garimella PS, Ambrosius WT, Cheung AK, Gren LH, Neyra JA, Punzi H, Raphael KL, Shlipak MG, Ix JH. Distinct Dimensions of Kidney Health and Risk of Cardiovascular Disease, Heart Failure, and Mortality. Hypertension 2019; 74:872-879. [PMID: 31378102 PMCID: PMC6739187 DOI: 10.1161/hypertensionaha.119.13339] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 06/05/2019] [Indexed: 12/20/2022]
Abstract
Chronic kidney disease is a strong risk factor for cardiovascular disease (CVD), but clinical kidney measures (estimated glomerular filtration rate and albuminuria) do not fully reflect the multiple aspects of kidney tubules influencing cardiovascular health. Applied methods are needed to integrate numerous tubule biomarkers into useful prognostic scores. In SPRINT (Systolic Blood Pressure Intervention Trial) participants with chronic kidney disease at baseline (estimated glomerular filtration ratecr&cys <60 mL/minute per 1.73 m2), we measured 8 biomarkers from urine (α1M [α1M microglobulin], β2M [β2M microglobulin], umod [uromodulin], KIM-1 [kidney injury molecule-1], MCP-1 [monocyte chemoattractant protein-1], YKL-40 [chitinase-3-like protein-1], NGAL [neutrophil gelatinase-associated lipocalin], and IL-18 [interleukin 18]) and 2 biomarkers from serum (intact parathyroid hormone, iFGF-23 [intact fibroblast growth factor-23]). We used an unsupervised method, exploratory factor analysis, to create summary scores of tubule health dimensions. Adjusted Cox models evaluated each tubule score with CVD events, heart failure, and all-cause mortality. We examined CVD discrimination using Harrell C-statistic. Factor analysis of 10 biomarkers from 2376 SPRINT-chronic kidney disease participants identified 4 unique dimensions of tubular health: tubule injury/repair (NGAL, IL-18, YKL-40), tubule injury/fibrosis (KIM-1, MCP-1), tubule reabsorption (α1M, β2M), and tubular reserve/mineral metabolism (umod, intact parathyroid hormone, iFGF-23). After adjustment for CVD risk factors, estimated glomerular filtration rate, and albumin-to-creatinine ratio, 2 of the 4 tubule scores were associated with CVD (hazard ratio per SD; reabsorption, 1.21 [1.06-1.38]; reserve, 1.24 (1.08-1.38]), 1 with heart failure (reserve, 1.41 [1.13-1.74]), and none with mortality. Compared with a base model (C-statistic=0.674), adding estimated glomerular filtration rate and albumin-to-creatinine ratio improved the C-statistic (C=0.704; P=0.001); further adding tubule scores additionally improved the C-statistic (C=0.719; P=0.009). In the setting of chronic kidney disease, dimensions of tubule health quantified using factor analysis improved CVD discrimination beyond contemporary kidney measures. Clinical Trial Registration- URL: http://www.clinicaltrials.gov. Unique identifier: NCT01206062.
Collapse
Affiliation(s)
| | | | | | | | | | - Alfred K. Cheung
- University of Utah
- Veterans Affairs Salt Lake City Healthcare System
| | | | - Javier A. Neyra
- University of Texas Southwestern, Dallas
- University of Kentucky, Lexington
| | | | | | | | | |
Collapse
|
21
|
Yan J, Huo J, Li R, Jia Z, Song Y, Chen J, Zhang L. Benchmark dose estimation of urinary and blood cadmium as biomarkers of renal dysfunction among 40‐75‐year‐old non‐smoking women in rural areas of southwest China. J Appl Toxicol 2019; 39:1433-1443. [PMID: 31313336 DOI: 10.1002/jat.3829] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/03/2019] [Accepted: 05/05/2019] [Indexed: 01/08/2023]
Affiliation(s)
- Jiuming Yan
- West China School of Public Health and West China Fourth Hospital, West China School of Public Health and Healthy Food Evaluation Research Center, Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan ProvinceSichuan University Chengdu Sichuan China
| | - Jiao Huo
- West China School of Public Health and West China Fourth Hospital, West China School of Public Health and Healthy Food Evaluation Research Center, Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan ProvinceSichuan University Chengdu Sichuan China
| | - Renjia Li
- West China School of Public Health and West China Fourth Hospital, West China School of Public Health and Healthy Food Evaluation Research Center, Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan ProvinceSichuan University Chengdu Sichuan China
| | - Zhenchao Jia
- West China School of Public Health and West China Fourth Hospital, West China School of Public Health and Healthy Food Evaluation Research Center, Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan ProvinceSichuan University Chengdu Sichuan China
| | - Yang Song
- West China School of Public Health and West China Fourth Hospital, West China School of Public Health and Healthy Food Evaluation Research Center, Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan ProvinceSichuan University Chengdu Sichuan China
| | - Jinyao Chen
- West China School of Public Health and West China Fourth Hospital, West China School of Public Health and Healthy Food Evaluation Research Center, Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan ProvinceSichuan University Chengdu Sichuan China
| | - Lishi Zhang
- West China School of Public Health and West China Fourth Hospital, West China School of Public Health and Healthy Food Evaluation Research Center, Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan ProvinceSichuan University Chengdu Sichuan China
| |
Collapse
|
22
|
Serchenya T, Shcharbin D, Shyrochyna I, Sviridov O, Terekhova M, Dzmitruk V, Abashkin V, Apartsin E, Mignani S, Majoral JP, Ionov M, Bryszewska M. Immunoreactivity changes of human serum albumin and alpha-1-microglobulin induced by their interaction with dendrimers. Colloids Surf B Biointerfaces 2019; 179:226-232. [DOI: 10.1016/j.colsurfb.2019.03.065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/11/2019] [Accepted: 03/28/2019] [Indexed: 01/15/2023]
|
23
|
Wan X, Zhang L, Gu H, Wang S, Liu X. The Association of Serum hsCRP and Urinary Alpha1-Microglobulin in Patients with Type 2 Diabetes Mellitus. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6364390. [PMID: 31281843 PMCID: PMC6590668 DOI: 10.1155/2019/6364390] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/27/2019] [Indexed: 11/17/2022]
Abstract
This study aimed to investigate the association of serum hsCRP and urinary A1MG in patients with T2DM. Numerous investigations have proven that serum hypersensitive C-reactive protein (hsCRP) concentration in patients with type 2 diabetes mellitus (T2DM) is increased. Also, increased urinary alpha-1 microglobulin (A1MG) can be an early sign of renal damage, primarily on the proximal tubules in T2DM. Little information is available with respect to the associations of serum hsCRP levels and urinary A1MG in T2DM. A total of 520 patients with T2DM were recruited to participate in this study. Serum hsCRP and UA1MG (urinary alpha1-microglobulin to creatinine ratio), UACR (urinary microalbumin to creatinine ratio), UIGG (urinary immunoglobulin G to creatinine ratio), and UTRF (urinary transferrin to creatinine ratio) were obtained. The association of serum hsCRP level and each urinary protein parameter was analyzed by using the regression analysis, respectively. LnhsCRP was positively associated with the lnUA1MG in all three linear regression models (adjusted β in model 3=0.122, SE=0.027, P<0.001). Furthermore, the high hsCRP group (hsCRP > 3mg/L) was associated with increasing risk of high UA1MG (adjusted OR in model 3=1.610, 95% CI 1.037-2.499, P=0.034) by logistic regression. This study suggests that serum hsCRP levels independently associate with UA1MG in patients with T2DM. Further research is warranted to elucidate these interactions.
Collapse
Affiliation(s)
- Xiaohua Wan
- Department of Clinical Laboratory, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Lin Zhang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, 100730, China
- Beijing Diabetes Institute, Beijing, 100730, China
| | - Haitong Gu
- Department of Clinical Laboratory, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Shenglai Wang
- Department of Clinical Laboratory, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Xiangyi Liu
- Department of Clinical Laboratory, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| |
Collapse
|
24
|
Pre-exposure Prophylaxis With Tenofovir Disoproxil Fumarate/Emtricitabine and Kidney Tubular Dysfunction in HIV-Uninfected Individuals. J Acquir Immune Defic Syndr 2019; 78:169-174. [PMID: 29767638 PMCID: PMC6071417 DOI: 10.1097/qai.0000000000001654] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Pre-exposure prophylaxis (PrEP) with tenofovir disoproxil fumarate (TDF) and emtricitabine (FTC) is becoming increasingly adopted for HIV prevention. Tenofovir can cause proximal tubular damage and chronic kidney disease in HIV-infected persons, but little is known regarding its nephrotoxic potential among HIV-uninfected persons. In this study, we evaluated the effects of PrEP on urine levels of the following: α1-microglobulin (α1m), a marker of impaired tubular reabsorption; albuminuria, a measure of glomerular injury; and total proteinuria. SETTING The Iniciativa Profilaxis Pre-Exposicion (iPrEx) study randomized HIV-seronegative men and transgender women who have sex with men to oral TDF/FTC or placebo. The iPrEx open-label extension (iPrEx-OLE) study enrolled former PrEP trial participants to receive open-label TDF/FTC. METHODS A cross-sectional analysis compared urine biomarker levels by study arm in iPrEx (N = 100 treatment arm, N = 100 placebo arm). Then, urine biomarker levels were compared before and after PrEP initiation in 109 participants of iPrEx-OLE. RESULTS In iPrEx, there were no significant differences in urine α1m, albuminuria, or proteinuria by treatment arm. In iPrEx-OLE, after 24 weeks on PrEP, urine α1m and proteinuria increased by 21% [95% confidence interval (CI): 10 to 33] and 18% (95% CI: 8 to 28), respectively. The prevalence of detectable α1m increased from 44% to 65% (P < 0.001) and estimated glomerular filtration rate declined by 4 mL/min/1.73 m (P < 0.001). There was no significant change in albuminuria (6%; 95% CI: -7% to 20%). CONCLUSION PrEP with TDF/FTC was associated with a statistically significant rise in urine α1m and proteinuria after 6 months, suggesting that PrEP may result in subclinical tubule dysfunction.
Collapse
|
25
|
Treacy O, Brown NN, Dimeski G. Biochemical evaluation of kidney disease. Transl Androl Urol 2019; 8:S214-S223. [PMID: 31236339 PMCID: PMC6559936 DOI: 10.21037/tau.2018.10.02] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/08/2018] [Indexed: 12/17/2022] Open
Abstract
Different biochemical markers exist in both blood and urine for assessing renal function. Most of these biomarkers have advantages and limitations associated with their use, which is important to consider when ordering and utilising them in the clinical setting. The ideal marker should be able to detect acute kidney injury (AKI) at the onset and be used for the diagnosis and ongoing monitoring and management of kidney disease. The search for such a marker is ongoing, as all potential candidates thus far are associated with certain limitations. This article will attempt to compare and contrast established and emerging kidney disease markers.
Collapse
Affiliation(s)
- Oliver Treacy
- Department of Chemical Pathology, Pathology Queensland, Princess Alexandra Hospital Ipswich Road, Woolloongabba, Queensland, Australia
| | - Nigel N. Brown
- Department of Chemical Pathology, Pathology Queensland, Princess Alexandra Hospital Ipswich Road, Woolloongabba, Queensland, Australia
| | - Goce Dimeski
- Department of Chemical Pathology, Pathology Queensland, Princess Alexandra Hospital Ipswich Road, Woolloongabba, Queensland, Australia
- School of Medicine, University of Queensland, Brisbane, Australia
| |
Collapse
|
26
|
Charkoftaki G, Wang Y, McAndrews M, Bruford EA, Thompson DC, Vasiliou V, Nebert DW. Update on the human and mouse lipocalin (LCN) gene family, including evidence the mouse Mup cluster is result of an "evolutionary bloom". Hum Genomics 2019; 13:11. [PMID: 30782214 PMCID: PMC6381713 DOI: 10.1186/s40246-019-0191-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/17/2019] [Indexed: 12/12/2022] Open
Abstract
Lipocalins (LCNs) are members of a family of evolutionarily conserved genes present in all kingdoms of life. There are 19 LCN-like genes in the human genome, and 45 Lcn-like genes in the mouse genome, which include 22 major urinary protein (Mup) genes. The Mup genes, plus 29 of 30 Mup-ps pseudogenes, are all located together on chromosome (Chr) 4; evidence points to an “evolutionary bloom” that resulted in this Mup cluster in mouse, syntenic to the human Chr 9q32 locus at which a single MUPP pseudogene is located. LCNs play important roles in physiological processes by binding and transporting small hydrophobic molecules —such as steroid hormones, odorants, retinoids, and lipids—in plasma and other body fluids. LCNs are extensively used in clinical practice as biochemical markers. LCN-like proteins (18–40 kDa) have the characteristic eight β-strands creating a barrel structure that houses the binding-site; LCNs are synthesized in the liver as well as various secretory tissues. In rodents, MUPs are involved in communication of information in urine-derived scent marks, serving as signatures of individual identity, or as kairomones (to elicit fear behavior). MUPs also participate in regulation of glucose and lipid metabolism via a mechanism not well understood. Although much has been learned about LCNs and MUPs in recent years, more research is necessary to allow better understanding of their physiological functions, as well as their involvement in clinical disorders.
Collapse
Affiliation(s)
- Georgia Charkoftaki
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, 06520-8034, USA
| | - Yewei Wang
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, 06520-8034, USA
| | - Monica McAndrews
- Mouse Genome Informatics, The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Elspeth A Bruford
- HUGO Gene Nomenclature Committee, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - David C Thompson
- Department of Clinical Pharmacy, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, USA
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, 06520-8034, USA.
| | - Daniel W Nebert
- Department of Environmental Health and Center for Environmental Genetics; Department of Pediatrics and Molecular and Developmental Biology, Cincinnati Children's Research Center, University Cincinnati Medical Center, Cincinnati, OH, 45267, USA
| |
Collapse
|
27
|
Hakuno D, Kimura M, Ito S, Satoh J, Nakashima Y, Horie T, Kuwabara Y, Nishiga M, Ide Y, Baba O, Nishi H, Nakao T, Nishino T, Nakazeki F, Koyama S, Hanada R, Randolph RR, Endo J, Kimura T, Ono K. Hepatokine α1-Microglobulin Signaling Exacerbates Inflammation and Disturbs Fibrotic Repair in Mouse Myocardial Infarction. Sci Rep 2018; 8:16749. [PMID: 30425314 PMCID: PMC6233179 DOI: 10.1038/s41598-018-35194-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 10/30/2018] [Indexed: 12/31/2022] Open
Abstract
Acute cardiac rupture and adverse left ventricular (LV) remodeling causing heart failure are serious complications of acute myocardial infarction (MI). While cardio-hepatic interactions have been recognized, their role in MI remains unknown. We treated cultured cardiomyocytes with conditioned media from various cell types and analyzed the media by mass spectrometry to identify α1-microglobulin (AM) as an Akt-activating hepatokine. In mouse MI model, AM protein transiently distributed in the infarct and border zones during the acute phase, reflecting infiltration of AM-bound macrophages. AM stimulation activated Akt, NFκB, and ERK signaling and enhanced inflammation as well as macrophage migration and polarization, while inhibited fibrogenesis-related mRNA expression in cultured macrophages and cardiac fibroblasts. Intramyocardial AM administration exacerbated macrophage infiltration, inflammation, and matrix metalloproteinase 9 mRNA expression in the infarct and border zones, whereas disturbed fibrotic repair, then provoked acute cardiac rupture in MI. Shotgun proteomics and lipid pull-down analysis found that AM partly binds to phosphatidic acid (PA) for its signaling and function. Furthermore, systemic delivery of a selective inhibitor of diacylglycerol kinase α-mediated PA synthesis notably reduced macrophage infiltration, inflammation, matrix metalloproteinase activity, and adverse LV remodeling in MI. Therefore, targeting AM signaling could be a novel pharmacological option to mitigate adverse LV remodeling in MI.
Collapse
Affiliation(s)
- Daihiko Hakuno
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Masahiro Kimura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Shinji Ito
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Junko Satoh
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yasuhiro Nakashima
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takahiro Horie
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yasuhide Kuwabara
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Masataka Nishiga
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yuya Ide
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Osamu Baba
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hitoo Nishi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tetsushi Nakao
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tomohiro Nishino
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Fumiko Nakazeki
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Satoshi Koyama
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ritsuko Hanada
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ruiz R Randolph
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Jin Endo
- Cardiovascular Division, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Koh Ono
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
28
|
Kozyraki R, Cases O. Cubilin, the Intrinsic Factor-Vitamin B12 Receptor in Development and Disease. Curr Med Chem 2018; 27:3123-3150. [PMID: 30295181 DOI: 10.2174/0929867325666181008143945] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 08/11/2018] [Accepted: 08/21/2018] [Indexed: 12/29/2022]
Abstract
Gp280/Intrinsic factor-vitamin B12 receptor/Cubilin (CUBN) is a large endocytic receptor serving multiple functions in vitamin B12 homeostasis, renal reabsorption of protein or toxic substances including albumin, vitamin D-binding protein or cadmium. Cubilin is a peripheral membrane protein consisting of 8 Epidermal Growth Factor (EGF)-like repeats and 27 CUB (defined as Complement C1r/C1s, Uegf, BMP1) domains. This structurally unique protein interacts with at least two molecular partners, Amnionless (AMN) and Lrp2/Megalin. AMN is involved in appropriate plasma membrane transport of Cubilin whereas Lrp2 is essential for efficient internalization of Cubilin and its ligands. Observations gleaned from animal models with Cubn deficiency or human diseases demonstrate the importance of this protein. In this review addressed to basic research and medical scientists, we summarize currently available data on Cubilin and its implication in renal and intestinal biology. We also discuss the role of Cubilin as a modulator of Fgf8 signaling during embryonic development and propose that the Cubilin-Fgf8 interaction may be relevant in human pathology, including in cancer progression, heart or neural tube defects. We finally provide experimental elements suggesting that some aspects of Cubilin physiology might be relevant in drug design.
Collapse
Affiliation(s)
- Renata Kozyraki
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris-Diderot University, Paris, France
| | - Olivier Cases
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris-Diderot University, Paris, France
| |
Collapse
|
29
|
Sekikawa S, Onda T, Miura N, Nomura T, Takano N, Shibahara T, Honda K. Underexpression of α-1-microglobulin/bikunin precursor predicts a poor prognosis in oral squamous cell carcinoma. Int J Oncol 2018; 53:2605-2614. [PMID: 30272341 DOI: 10.3892/ijo.2018.4581] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 08/24/2018] [Indexed: 11/06/2022] Open
Abstract
In the present study, in order to identify novel diagnostic biomarkers for the malignant behavior of oral squamous cell carcinoma (OSCC), we determined the proteomic profiles of several OSCC cell lines and keratinocytes by two-dimensional fluorescence difference gel electrophoresis and liquid chromatography tandem mass spectrometry. The protein expression level of α-1-microglobulin/bikunin precursor (AMBP) was found to be significantly lower in the OSCC cell lines than in the keratinocytes, and a significant decrease in AMBP mRNA expression was confirmed in the OSCC cell lines by RT-qPCR. To investigate the biological function of AMBP in OSCC, the cells were transiently transfected with an AMBP overexpression vector; the AMBP-overexpressing cells exhibited a significantly decreased invasion and migration in comparison to the mock-transfected control cells, although no significant changes in cell proliferation were observed. Immunohistochemistry revealed that the underexpression of AMBP was significantly associated with a high metastatic potential to cervical lymph nodes and a poor overall survival. Thus, the expression of AMBP is an independent predictive factor of cervical lymph node metastasis and a prognostic factor of overall survival, and it is involved in both cell invasion and metastasis in cervical lymph nodes in OSCC.
Collapse
Affiliation(s)
- Shoichi Sekikawa
- Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Chiba 261-8502, Japan
| | - Takeshi Onda
- Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Chiba 261-8502, Japan
| | - Nami Miura
- Department of Biomarkers for Early Detection of Cancer, National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | - Takeshi Nomura
- Department of Oral Medicine, Oral and Maxillofacial Surgery, Tokyo Dental College, Chiba 272-8513, Japan
| | - Nobuo Takano
- Oral Cancer Center, Tokyo Dental College, Chiba 272-8513, Japan
| | - Takahiko Shibahara
- Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Chiba 261-8502, Japan
| | - Kazufumi Honda
- Department of Biomarkers for Early Detection of Cancer, National Cancer Center Research Institute, Tokyo 104-0045, Japan
| |
Collapse
|
30
|
Jotwani V, Katz R, Ix JH, Gutiérrez OM, Bennett M, Parikh CR, Cummings SR, Sarnak MJ, Shlipak MG. Urinary Biomarkers of Kidney Tubular Damage and Risk of Cardiovascular Disease and Mortality in Elders. Am J Kidney Dis 2018; 72:205-213. [PMID: 29602632 DOI: 10.1053/j.ajkd.2017.12.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 12/23/2017] [Indexed: 01/07/2023]
Abstract
RATIONALE & OBJECTIVE Novel urinary biomarkers have enabled earlier detection of kidney tubular damage, but their prognostic value for adverse cardiovascular outcomes is uncertain. We hypothesized that tubular damage, measured by urine α1-microglobulin (A1M), amino-terminal propeptide of type III procollagen (PIIINP), and neutrophil gelatinase-associated lipocalin (NGAL), would be associated with higher risks for cardiovascular events and mortality among elders. STUDY DESIGN Case-cohort study. SETTING & PARTICIPANTS This study included a randomly selected subcohort (n=502), cardiovascular disease (CVD) cases (n=245), and heart failure cases (n=220) from the Health, Aging, and Body Composition (Health ABC) Study. PREDICTORS Baseline urine A1M, PIIINP, and NGAL concentrations. OUTCOMES Incident CVD, heart failure, and all-cause mortality. ANALYTICAL APPROACH Cox proportional hazards models were used to evaluate biomarker associations with each outcome. RESULTS At baseline, mean age was 74 years and estimated glomerular filtration rate was 73mL/min/1.73m2. After adjustment for demographics, estimated glomerular filtration rate, albumin-creatinine ratio, and other cardiovascular risk factors, each doubling in biomarker concentration was associated with the following adjusted HRs for CVD: A1M, 1.51 (95% CI, 1.16-1.96); PIIINP, 1.21 (95% CI, 1.00-1.46); and NGAL, 1.12 (95% CI, 1.05-1.20). There were 248 deaths in the subcohort during a median follow-up of 12.4 years. Adjusted associations of each biomarker (HR per doubling) with all-cause mortality were: A1M, 1.29 (95% CI, 1.10-1.51); PIIINP, 1.05 (95%, 0.94-1.18); and NGAL, 1.07 (95% CI, 1.02-1.12). Biomarker concentrations did not have statistically significant associations with heart failure after multivariable adjustment. LIMITATIONS Urine biomarkers were measured at a single time point; no validation cohort available. CONCLUSIONS Kidney tubular damage is an independent risk factor for CVD and death among elders. Future studies should investigate mechanisms by which kidney tubular damage may adversely affect cardiovascular risk.
Collapse
Affiliation(s)
- Vasantha Jotwani
- Division of Nephrology, Department of Medicine, San Francisco VA Medical Center, San Francisco, CA; Kidney Health Research Collaborative, San Francisco VA Medical Center and University of California, San Francisco, CA.
| | - Ronit Katz
- Kidney Research Institute, University of Washington, Seattle, WA
| | - Joachim H Ix
- Division of Nephrology-Hypertension, Department of Medicine, University of California, San Diego, CA; Nephrology Section, Veterans Affairs San Diego Healthcare System, San Diego, CA
| | - Orlando M Gutiérrez
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, AL
| | - Michael Bennett
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Chirag R Parikh
- Section of Nephrology, Department of Medicine, Yale University, New Haven, CT; Program of Applied Translational Research, Yale University, New Haven, CT
| | - Steven R Cummings
- California Pacific Medical Center Research Institute, San Francisco, CA
| | - Mark J Sarnak
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, MA
| | - Michael G Shlipak
- Division of Nephrology, Department of Medicine, San Francisco VA Medical Center, San Francisco, CA; Kidney Health Research Collaborative, San Francisco VA Medical Center and University of California, San Francisco, CA
| |
Collapse
|
31
|
Park M, Katz R, Shlipak MG, Weiner D, Tracy R, Jotwani V, Hughes-Austin J, Gabbai F, Hsu CY, Pfeffer M, Bansal N, Bostom A, Gutierrez O, Sarnak M, Levey A, Ix JH. Urinary Markers of Fibrosis and Risk of Cardiovascular Events and Death in Kidney Transplant Recipients: The FAVORIT Trial. Am J Transplant 2017; 17:2640-2649. [PMID: 28371433 PMCID: PMC5620109 DOI: 10.1111/ajt.14284] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 03/13/2017] [Accepted: 03/18/2017] [Indexed: 01/25/2023]
Abstract
Cardiovascular risk remains high in kidney transplant recipients (KTRs) despite improved kidney function after transplant. Urinary markers of kidney fibrosis and injury may help to reveal mechanisms of this risk. In a case-cohort study among stable KTRs who participated in the FAVORIT trial, we measured four urinary proteins known to correlate with kidney tubulointerstitial fibrosis on biopsy (urine alpha 1 microglobulin [α1m], monocyte chemoattractant protein-1 [MCP-1], procollagen type I [PINP] and type III [PIIINP] N-terminal amino peptide) and evaluated associations with cardiovascular disease (CVD) events (n = 300) and death (n = 371). In adjusted models, higher urine α1m (hazard ratio [HR] per doubling of biomarker 1.40 [95% confidence interval [CI] 1.21, 1.62]), MCP-1 (HR 1.18 [1.03, 1.36]), and PINP (HR 1.13 [95% CI 1.03, 1.23]) were associated with CVD events. These three markers were also associated with death (HR per doubling α1m 1.51 [95% CI 1.32, 1.72]; MCP-1 1.31 [95% CI 1.13, 1.51]; PINP 1.11 [95% CI 1.03, 1.20]). Higher concentrations of urine α1m, MCP-1, and PINP may identify KTRs at higher risk for CVD events and death. These markers may identify a systemic process of fibrosis involving both the kidney and cardiovascular system, and give new insights into mechanisms linking the kidney with CVD.
Collapse
Affiliation(s)
- M Park
- Division of Nephrology, Department of Medicine, University of California San Francisco, San Francisco, California,Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California
| | - R Katz
- Kidney Research Institute, Division of Nephrology, University of Washington, Seattle, Washington
| | - M G Shlipak
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California,General Internal Medicine Section, San Francisco Veterans Affairs Hospital, San Francisco, California,Division of General Internal Medicine, Department of Medicine, University of California San Francisco, San Francisco, California
| | - D Weiner
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - R Tracy
- Department of Pathology, University of Vermont, Burlington, Vermont
| | - V Jotwani
- Division of Nephrology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - J Hughes-Austin
- Division of Preventive Medicine, Department of Preventive Medicine and Public Health, University of California San Diego, San Diego, California
| | - F Gabbai
- Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, San Diego, California,Nephrology Section, Veterans Affairs San Diego Healthcare System, San Diego, California
| | - CY Hsu
- Division of Nephrology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - M Pfeffer
- Division of Cardiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - N Bansal
- Kidney Research Institute, Division of Nephrology, University of Washington, Seattle, Washington
| | - A Bostom
- Rhode Island Hospital, Providence, Rhode Island
| | - O Gutierrez
- Departments of Medicine and Epidemiology, University of Alabama at Birmingham, Birmingham, AL
| | - M Sarnak
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - A Levey
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - J H Ix
- Division of Preventive Medicine, Department of Preventive Medicine and Public Health, University of California San Diego, San Diego, California,Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, San Diego, California,Nephrology Section, Veterans Affairs San Diego Healthcare System, San Diego, California
| |
Collapse
|
32
|
Abstract
Alpha 1 microglobulin is a low molecular weight heme binding antioxidant protein with interesting, and potentially important, clinical applications. However, much remains to be learned about its in vivo effects. This invited review raises a number of physiologic issues regarding this compound as it pertains to clinical use.
Collapse
Affiliation(s)
- Richard A Zager
- Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
33
|
Codsi E, Garovic VD, Gonzalez-Suarez ML, Milic N, Borowski KS, Rose CH, Davies NP, Kashani KB, Lieske JC, White WM. Longitudinal characterization of renal proximal tubular markers in normotensive and preeclamptic pregnancies. Am J Physiol Regul Integr Comp Physiol 2017; 312:R773-R778. [DOI: 10.1152/ajpregu.00509.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 03/29/2017] [Accepted: 04/18/2017] [Indexed: 01/10/2023]
Abstract
Glomerular damage is common in preeclampsia (PE), but the extent and etiology of tubular injury are not well understood. The aim of this study was to evaluate tubular injury in patients with PE and to assess whether it predates clinical disease. We performed a prospective cohort study of 315 pregnant women who provided urine samples at the end of the second trimester and at delivery. This analysis included women who developed PE ( n = 15), gestational hypertension (GH; n = 14), and normotensive controls (NC; n = 44). Urinary markers of tubular injury, α1-microglobulin (A1M), retinol-binding protein (RBP), kidney-injury molecule-1 (KIM1), complement C5b-9, tissue inhibitor metalloproteinase-2 (TIMP-2), and insulin-like growth factor binding protein-7 (IGFBP-7) were measured by enzyme-linked immunosorbent assay (ELISA) and reported in relation to urine creatinine concentration. Second-trimester concentrations of all markers were similar among groups. At delivery, A1M concentrations were higher in the PE group than in the GH and NC groups as an A1M/creatinine ratio >13 (66.7, 8.3, and 35%, respectively, P = 0.01). Concentrations of C5b-9 were higher in the PE group than in the GH and NC groups (medians 9.85, 0.05, and 0.28 ng/mg, respectively, P = 0.003). KIM1, RBP, TIMP-2, and IGFBP-7 concentrations did not differ among groups at delivery. In conclusion, proximal tubular dysfunction, as assessed by A1M and C5b-9, developed during the interval between the end of the second trimester and delivery in patients with PE. However, this was not matched by abnormalities in markers previously associated with tubular cell injury (KIM-1, IGFBP-7, and TIMP-2).
Collapse
Affiliation(s)
- Elisabeth Codsi
- Department of Maternal Fetal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Vesna D. Garovic
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota; and
| | - Maria L. Gonzalez-Suarez
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota; and
| | - Natasa Milic
- Department of Medical Statistics and Informatics, University of Belgrade, Belgrade, Serbia
| | - Kristi S. Borowski
- Department of Maternal Fetal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Carl H. Rose
- Department of Maternal Fetal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Norman P. Davies
- Department of Maternal Fetal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Kianoush B. Kashani
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota; and
| | - John C. Lieske
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota; and
| | - Wendy M. White
- Department of Maternal Fetal Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
34
|
Ix JH, Katz R, Bansal N, Foster M, Weiner DE, Tracy R, Jotwani V, Hughes-Austin J, McKay D, Gabbai F, Hsu CY, Bostom A, Levey AS, Shlipak MG. Urine Fibrosis Markers and Risk of Allograft Failure in Kidney Transplant Recipients: A Case-Cohort Ancillary Study of the FAVORIT Trial. Am J Kidney Dis 2017; 69:410-419. [PMID: 28024930 PMCID: PMC7321838 DOI: 10.1053/j.ajkd.2016.10.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 10/05/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND Kidney tubulointerstitial fibrosis marks risk for allograft failure in kidney transplant recipients, but is poorly captured by estimated glomerular filtration rate (eGFR) or urine albumin-creatinine ratio (ACR). Whether urinary markers of tubulointerstitial fibrosis can noninvasively identify risk for allograft failure above and beyond eGFR and ACR is unknown. STUDY DESIGN Case-cohort study. SETTING & PARTICIPANTS The FAVORIT (Folic Acid for Vascular Outcome Reduction in Transplantation) Trial was a randomized double-blind trial testing vitamin therapy to lower homocysteine levels in stable kidney transplant recipients. We selected a subset of participants at random (n=491) and all individuals with allograft failure during follow-up (cases; n=257). PREDICTOR Using spot urine specimens from the baseline visit, we measured 4 urinary proteins known to correlate with tubulointerstitial fibrosis on biopsy (urine α1-microglobulin [A1M], monocyte chemoattractant protein 1 [MCP-1], and procollagen type III and type I amino-terminal amino pro-peptide). OUTCOME Death-censored allograft failure. RESULTS In models adjusted for demographics, chronic kidney disease risk factors, eGFR, and ACR, higher concentrations of urine A1M (HR per doubling, 1.73; 95% CI, 1.43-2.08) and MCP-1 (HR per doubling, 1.60; 95% CI, 1.32-1.93) were strongly associated with allograft failure. When additionally adjusted for concentrations of other urine fibrosis and several urine injury markers, urine A1M (HR per doubling, 1.76; 95% CI, 1.27-2.44]) and MCP-1 levels (HR per doubling, 1.49; 95% CI, 1.17-1.89) remained associated with allograft failure. Urine procollagen type III and type I levels were not associated with allograft failure. LIMITATIONS We lack kidney biopsy data, BK titers, and HLA antibody status. CONCLUSIONS Urine measurement of tubulointerstitial fibrosis may provide a noninvasive method to identify kidney transplant recipients at higher risk for future allograft failure, above and beyond eGFR and urine ACR.
Collapse
Affiliation(s)
- Joachim H Ix
- Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, San Diego, CA; Nephrology Section, Veterans Affairs San Diego Healthcare System, San Diego, CA; Division of Preventive Medicine, Department of Family Medicine and Public Health, University of California San Diego, San Diego, CA.
| | - Ronit Katz
- Kidney Research Institute, Division of Nephrology, University of Washington, Seattle, WA
| | - Nisha Bansal
- Kidney Research Institute, Division of Nephrology, University of Washington, Seattle, WA
| | - Meredith Foster
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, MA
| | - Daniel E Weiner
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, MA
| | - Russell Tracy
- Department of Pathology, University of Vermont, Burlington, VT
| | - Vasantha Jotwani
- Division of Nephrology, Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Jan Hughes-Austin
- Department of Orthopedic Surgery, University of California San Diego, San Diego, CA
| | - Dianne McKay
- Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, San Diego, CA
| | - Francis Gabbai
- Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, San Diego, CA; Nephrology Section, Veterans Affairs San Diego Healthcare System, San Diego, CA
| | - Chi-Yuan Hsu
- Division of Nephrology, Department of Medicine, University of California San Francisco, San Francisco, CA
| | | | - Andrew S Levey
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, MA
| | - Michael G Shlipak
- General Internal Medicine Section, San Francisco Veterans Affairs Hospital, San Francisco, CA; Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA; Division of General Internal Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA
| |
Collapse
|
35
|
Zager RA, Johnson ACM, Frostad K. An evaluation of the antioxidant protein α1-microglobulin as a renal tubular cytoprotectant. Am J Physiol Renal Physiol 2016; 311:F640-51. [DOI: 10.1152/ajprenal.00264.2016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 05/04/2016] [Indexed: 11/22/2022] Open
Abstract
α1-Microglobulin (A1M) is a low-molecular-weight heme-binding antioxidant protein that is readily filtered by the glomerulus and reabsorbed by proximal tubules. Given these properties, recombinant A1M (rA1M) has been proposed as a renal antioxidant and therapeutic agent. However, little direct evidence to support this hypothesis exists. Hence, we have sought “proof of concept” in this regard. Cultured proximal tubule (HK-2) cells or isolated mouse proximal tubule segments were challenged with a variety of prooxidant insults: 1) hemin, 2) myoglobin; 3) “catalytic” iron, 4) H2O2/Fenton reagents, 5) a Ca2+ ionophore, 6) antimycin A, or 7) hypoxia (with or without rA1M treatment). HK-2 injury was gauged by the percent lactate dehydrogenase release and 4,5-(dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide uptake. In vivo protection was sought in rA1M-treated mice subjected to 1) graded myohemoglobinura (2, 4, 8, or 9 ml/kg glycerol injection), 2) purified myoglobinemia/uria, or 3) endotoxemia. In vivo injury was assessed by blood urea nitrogen, creatinine, and the expression of redox-sensitive genes (heme oxygenase-1, neutrophil gelatinase-associated lipocalin, and monocyte chemoattractant protein-1 mRNAs). Although rA1M totally blocked in vitro hemin toxicity, equimolar albumin (another heme binder) or 10% serum induced equal protection. rA1M failed to mitigate any nonhemin forms of either in vitro or in vivo injury. A1M appeared to be rapidly degraded within proximal tubules (by Western blot analysis). Surprisingly, rA1M exerted select injury-promoting effects (increased in vitro catalytic iron/antimycin toxicities and increased in vivo monocyte chemoattractant protein-1/neutrophil gelatinase-associated lipocalin mRNA expression after glycerol or endotoxin injection). We conclude that rA1M has questionable utility as a renal antioxidant/cytoprotective agent, particularly in the presence of larger amounts of competitive free heme (e.g., albumin) binders.
Collapse
Affiliation(s)
- Richard A. Zager
- Fred Hutchinson Cancer Center, Seattle, Washington; and
- University of Washington, Seattle Washington
| | | | | |
Collapse
|
36
|
Allen S, McBride WT, Young IS, MacGowan SW, McMurray TJ, Prabhu S, Penugonda SP, Armstrong MA. A clinical, renal and immunological assessment of Surface Modifying Additive Treated (SMART™) cardiopulmonary bypass circuits. Perfusion 2016; 20:255-62. [PMID: 16231621 DOI: 10.1191/0267659105pf815oa] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Biocompatible cardiopulmonary bypass (CPB) circuits aim to reduce contact activation and its physiological consequences. We investigated the hypothesis that use of Surface Modifying Additive (SMA)-treated circuits (Sorin Group Ltd) compared with non-SMA circuits would be associated with preservation of blood pressure during CPB and modulation of perioperative subclinical renal function (urinary α-1-microglobulin (α-1-m)) and plasma and urinary cytokine changes. In a study of low-risk CABG patients ( n=40), randomized to SMA ( n=20) versus non-SMA circuits ( n=20), we found better preserved blood pressure at CPB initiation in SMA patients (p <0.05), particularly in ACE-inhibited SMA patients ( n=11) versus ACE-inhibited non-SMA patients ( n=10) (p <0.05). Plasma anti-inflammatory IL-10, as well as urinary α-1-m, were elevated 48 hours postoperatively (p <0.05). SMA patients also had lower blood loss (p <0.05). SMA circuits have some clinical benefit, especially in ACE-inhibited patients.
Collapse
Affiliation(s)
- Stephen Allen
- Department of Anaesthetics and Intensive Care Medicine, The Queen's University of Belfast, Belfast, Northern Ireland.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Jotwani V, Scherzer R, Estrella MM, Jacobson LP, Witt MD, Palella FJ, Macatangay B, Bennett M, Parikh CR, Ix JH, Shlipak MG. HIV Infection, Tenofovir, and Urine α1-Microglobulin: A Cross-sectional Analysis in the Multicenter AIDS Cohort Study. Am J Kidney Dis 2016; 68:571-581. [PMID: 27287300 DOI: 10.1053/j.ajkd.2016.03.430] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 03/29/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Tenofovir disoproxil fumarate (TDF) can cause proximal tubular damage and chronic kidney disease in human immunodeficiency virus (HIV)-infected individuals. Urine α1-microglobulin (A1M), a low-molecular-weight protein indicative of proximal tubular dysfunction, may enable earlier detection of TDF-associated tubular toxicity. STUDY DESIGN Cross-sectional. SETTING & PARTICIPANTS 883 HIV-infected and 350 -uninfected men enrolled in the Multicenter AIDS Cohort Study. PREDICTORS HIV infection and TDF exposure. OUTCOME Urine A1M level. RESULTS Urine A1M was detectable in 737 (83%) HIV-infected and 202 (58%) -uninfected men (P<0.001). Among HIV-infected participants, 573 (65%) were current TDF users and 112 (13%) were past TDF users. After multivariable adjustment including demographics, traditional kidney disease risk factors, and estimated glomerular filtration rate, HIV infection was associated with 136% (95% CI, 104%-173%) higher urine A1M levels and 1.5-fold (95% CI, 1.3- to 1.6-fold) prevalence of detectable A1M. When participants were stratified by TDF exposure, HIV infection was associated with higher adjusted A1M levels, by 164% (95% CI, 127%-208%) among current users, 124% (95% CI, 78%-183%) among past users, and 76% (95% CI, 45%-115%) among never users. Among HIV-infected participants, each year of cumulative TDF exposure was associated with 7.6% (95% CI, 5.4%-9.9%) higher A1M levels in fully adjusted models, a 4-fold effect size relative to advancing age (1.8% [95% CI, 0.9%-2.7%] per year). Each year since TDF treatment discontinuation was associated with 4.9% (95% CI, -9.4%--0.2%) lower A1M levels among past users. LIMITATIONS Results may not be generalizable to women. CONCLUSIONS HIV-infected men had higher urine A1M levels compared with HIV-uninfected men. Among HIV-infected men, cumulative TDF exposure was associated with incrementally higher A1M levels, whereas time since TDF treatment discontinuation was associated with progressively lower A1M levels. Urine A1M appears to be a promising biomarker for detecting and monitoring TDF-associated tubular toxicity.
Collapse
Affiliation(s)
- Vasantha Jotwani
- Department of Medicine, San Francisco VA Medical Center, San Francisco, CA; Department of Epidemiology and Biostatistics, University of California, San Francisco, CA.
| | - Rebecca Scherzer
- Department of Medicine, San Francisco VA Medical Center, San Francisco, CA; Department of Epidemiology and Biostatistics, University of California, San Francisco, CA
| | | | - Lisa P Jacobson
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Mallory D Witt
- Division of HIV Medicine, Department of Medicine, Harbor-UCLA Medical Center and the Los Angeles Biomedical Research Institute at Harbor-UCLA, Torrance, CA
| | - Frank J Palella
- Division of Infectious Diseases, Department of Medicine, Northwestern University, Chicago, IL
| | - Bernard Macatangay
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Michael Bennett
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Chirag R Parikh
- Section of Nephrology, Department of Medicine, Yale University, New Haven, CT; Program of Applied Translational Research, Yale University, New Haven, CT
| | - Joachim H Ix
- Division of Nephrology-Hypertension, Department of Medicine, University of California, San Diego, CA; Nephrology Section, Veterans Affairs San Diego Healthcare System, San Diego, CA
| | - Michael G Shlipak
- Department of Medicine, San Francisco VA Medical Center, San Francisco, CA; Kidney Health Research Collaborative, San Francisco VA Medical Center and University of California, San Francisco, CA
| |
Collapse
|
38
|
di Masi A, Trezza V, Leboffe L, Ascenzi P. Human plasma lipocalins and serum albumin: Plasma alternative carriers? J Control Release 2016; 228:191-205. [PMID: 26951925 DOI: 10.1016/j.jconrel.2016.02.049] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 02/23/2016] [Accepted: 02/24/2016] [Indexed: 01/14/2023]
Abstract
Lipocalins are an evolutionarily conserved family of proteins that bind and transport a variety of exogenous and endogenous ligands. Lipocalins share a conserved eight anti-parallel β-sheet structure. Among the different lipocalins identified in humans, α-1-acid glycoprotein (AGP), apolipoprotein D (apoD), apolipoprotein M (apoM), α1-microglobulin (α1-m) and retinol-binding protein (RBP) are plasma proteins. In particular, AGP is the most important transporter for basic and neutral drugs, apoD, apoM, and RBP mainly bind endogenous molecules such as progesterone, pregnenolone, bilirubin, sphingosine-1-phosphate, and retinol, while α1-m binds the heme. Human serum albumin (HSA) is a monomeric all-α protein that binds endogenous and exogenous molecules like fatty acids, heme, and acidic drugs. Changes in the plasmatic levels of lipocalins and HSA are responsible for the onset of pathological conditions associated with an altered drug transport and delivery. This, however, does not necessary result in potential adverse effects in patients because many drugs can bind both HSA and lipocalins, and therefore mutual compensatory binding mechanisms can be hypothesized. Here, molecular and clinical aspects of ligand transport by plasma lipocalins and HSA are reviewed, with special attention to their role as alterative carriers in health and disease.
Collapse
Affiliation(s)
- Alessandra di Masi
- Dipartimento di Scienze, Università Roma Tre, Viale Marconi 446, I-00146 Roma, Italy; Istituto Nazionale di Biostrutture e Biosistemi, Via delle Medaglie d'Oro 305, I-00136 Roma, Italy.
| | - Viviana Trezza
- Dipartimento di Scienze, Università Roma Tre, Viale Marconi 446, I-00146 Roma, Italy
| | - Loris Leboffe
- Dipartimento di Scienze, Università Roma Tre, Viale Marconi 446, I-00146 Roma, Italy; Istituto Nazionale di Biostrutture e Biosistemi, Via delle Medaglie d'Oro 305, I-00136 Roma, Italy
| | - Paolo Ascenzi
- Istituto Nazionale di Biostrutture e Biosistemi, Via delle Medaglie d'Oro 305, I-00136 Roma, Italy; Laboratorio Interdipartimentale di Microscopia Elettronica, Università Roma Tre, Via della Vasca Navale 79, I-00146 Roma, Italy
| |
Collapse
|
39
|
Umbro I, Gentile G, Tinti F, Muiesan P, Mitterhofer AP. Recent advances in pathophysiology and biomarkers of sepsis-induced acute kidney injury. J Infect 2015; 72:131-42. [PMID: 26702738 DOI: 10.1016/j.jinf.2015.11.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 11/24/2015] [Accepted: 11/28/2015] [Indexed: 12/26/2022]
Abstract
Sepsis is a complex clinical syndrome characterized by a systemic inflammatory response to an infective insult. This process often leads to widespread tissue injury and multiple organ dysfunction. In particular, the development of acute kidney injury (AKI) is one of the most frequent complications, which increases the complexity and cost of care, and is an independent risk factor for mortality. Previous suggestions highlighting systemic hypotension, renal vasoconstriction and ischaemia-reperfusion injury as the primary pathophysiological mechanisms involved in sepsis-induced AKI have been challenged. Recently it has been shown that sepsis-induced AKI occurs in the setting of microvascular dysfunction with release of microparticles, inflammation and energetic adaptation of highly metabolic organs to cellular stress. The intolerable high mortality rate associated with sepsis-induced AKI is partially explained by an incomplete understanding of its pathophysiology and a delay in diagnosis. The aim of this review is to focus on advances in understanding the sepsis pathophysiology, with particular attention to the fundamental mechanisms of sepsis-induced AKI and the potential diagnostic and prognostic markers involved.
Collapse
Affiliation(s)
- Ilaria Umbro
- The Liver Unit, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston, B15 2GW Birmingham, United Kingdom; Department of Clinical Medicine, Nephrology and Dialysis B, Sapienza University of Rome, Viale dell'Università 37, 00185 Rome, Italy.
| | - Giuseppe Gentile
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Via Benevento 6, 00185 Rome, Italy.
| | - Francesca Tinti
- The Liver Unit, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston, B15 2GW Birmingham, United Kingdom; Department of Clinical Medicine, Nephrology and Dialysis B, Sapienza University of Rome, Viale dell'Università 37, 00185 Rome, Italy.
| | - Paolo Muiesan
- The Liver Unit, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston, B15 2GW Birmingham, United Kingdom.
| | - Anna Paola Mitterhofer
- Department of Clinical Medicine, Nephrology and Dialysis B, Sapienza University of Rome, Viale dell'Università 37, 00185 Rome, Italy.
| |
Collapse
|
40
|
Escobedo-Villarreal MM, Mercado-Moreira AB, Muñoz-Espinosa LE, Gamboa-Esparza M, Pérez-Rodríguez E, Cordero-Pérez P. [Urinary protein detection by iTRAQ® associated with renal transplant complications and its modification with therapy]. CIR CIR 2015; 83:393-401. [PMID: 26148981 DOI: 10.1016/j.circir.2015.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 03/23/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND After renal transplant, surgical, infection complications, as well as graft rejection may occur; early detection through non-invasive markers is the key to change therapy and avoid biopsy. OBJECTIVE The aime of the study is to determine urine protein profiles in patients undergoing renal transplant with complications and detect its variation when therapy is modified. MATERIAL AND METHODS Urine samples were collected from patients prior the transplant and various postoperative stages. Urinary protein profiles were obtained by peptide labeling using isobaric isotopes for relative quantification (iTRAQ(®)). RESULTS A total of 22 patients were included, of whom 12 developed post-transplant complication: 2 with graft rejection (one male and one female) and 10 (6 males and 4 females) in the group of post-transplant infections. Using iTRAQ(®) 15/345 and 28/113 proteins were identified and fulfilled the acceptance criteria, in graft rejection and post-transplant infections group, respectively. CONCLUSIONS Albumin was the only protein found in both groups, the remaining proteins were different. The 5 proteins with higher scores in graft rejection were: alpha-1-microglobulin, 5'-nucleotidase cytosolic III, retinol-binding protein 4, membrane protein palmitoylated 4, and serine carboxypeptidase, while post-transplant infections were: mitochondrial acetyl-coenzyme A synthetase, putative adenosyl homocysteinase 2, zinc finger protein GLIS1, putative protein FAM157B, and zinc finger protein 615. It remains to elucidate the involvement of each of these in patients with renal transplantation.
Collapse
Affiliation(s)
- Miguel Mariano Escobedo-Villarreal
- Servicio de Trasplantes, Departamento de Cirugía, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| | - Amanda Berenice Mercado-Moreira
- Unidad de Hígado, Departamento de Medicina Interna, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| | - Linda Elsa Muñoz-Espinosa
- Unidad de Hígado, Departamento de Medicina Interna, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| | - Mariana Gamboa-Esparza
- Unidad de Hígado, Departamento de Medicina Interna, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| | - Edelmiro Pérez-Rodríguez
- Servicio de Trasplantes, Departamento de Cirugía, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| | - Paula Cordero-Pérez
- Unidad de Hígado, Departamento de Medicina Interna, Hospital Universitario Dr. José Eleuterio González, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México.
| |
Collapse
|
41
|
Jotwani V, Shlipak MG, Scherzer R, Parekh RS, Kao WHL, Bennett M, Cohen MH, Nowicki M, Sharma A, Young M, Tien PC, Parikh CR, Estrella MM. APOL1 Genotype and Glomerular and Tubular Kidney Injury in Women With HIV. Am J Kidney Dis 2015; 65:889-98. [PMID: 25921719 PMCID: PMC4615696 DOI: 10.1053/j.ajkd.2015.02.329] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 02/15/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND APOL1 genotype is associated with advanced kidney disease in African Americans, but the pathogenic mechanisms are unclear. Here, associations of APOL1 genotype with urine biomarkers of glomerular and tubular injury and kidney function decline were evaluated. STUDY DESIGN Observational study. SETTING & PARTICIPANTS 431 human immunodeficiency virus (HIV)-infected African American women enrolled in Women's Interagency HIV Study (WIHS). PREDICTOR APOL1 genotype. OUTCOMES Albumin-creatinine ratio (ACR), 4 tubular injury biomarkers (interleukin 18 [IL-18], kidney injury molecule 1 [KIM-1], neutrophil gelatinase-associated lipocalin [NGAL], and α1-microglobulin [A1M]), and kidney function estimated using the CKD-EPI cystatin C equation. MEASUREMENTS Participants were genotyped for APOL1 single-nucleotide polymorphisms rs73885319 (G1 allele) and rs71785313 (G2 allele). Urine biomarkers were measured using stored samples from 1999-2000. Cystatin C was measured using serum collected at baseline and 4- and 8-year follow-ups. RESULTS At baseline, ACRs were higher among 47 women with 2 APOL1 risk alleles versus 384 women with 0/1 risk allele (median, 24 vs 11mg/g; P<0.001). Compared with women with 0/1 risk allele, women with 2 risk alleles had 104% higher ACRs (95% CI, 29-223mg/g) and 2-fold greater risk of ACR>30 (95% CI, 1.17-3.44) mg/g after multivariable adjustment. APOL1 genotype showed little association with urine IL-18:Cr ratio, KIM-1:Cr ratio, and NGAL:Cr ratio (estimates of -5% [95% CI, -24% to 18%], -20% [95% CI, -36% to -1%], and 10% [95% CI, -26% to 64%], respectively) or detectable urine A1M (prevalence ratio, 1.13; 95% CI, 0.65-1.97) in adjusted analyses. Compared with women with 0/1 allele, women with 2 risk alleles had faster eGFR decline, by 1.2 (95% CI, 0.2 to 2.2) mL/min/1.73m(2) per year, and 1.7- and 3.4-fold greater rates of incident chronic kidney disease (95% CI, 1.1 to 2.5) and 10% annual eGFR decline (95% CI, 1.7 to 6.7), respectively, with minimal attenuation after adjustment for glomerular and tubular injury biomarker levels. LIMITATIONS Results may not be generalizable to men. CONCLUSIONS Among HIV-infected African American women, APOL1-associated kidney injury appears to localize to the glomerulus, rather than the tubules.
Collapse
Affiliation(s)
- Vasantha Jotwani
- Department of Medicine, San Francisco VA Medical Center, San Francisco, CA; Department of Epidemiology and Biostatistics, University of California, San Francisco, CA.
| | - Michael G Shlipak
- Department of Medicine, San Francisco VA Medical Center, San Francisco, CA; Department of Epidemiology and Biostatistics, University of California, San Francisco, CA
| | - Rebecca Scherzer
- Department of Medicine, San Francisco VA Medical Center, San Francisco, CA; Department of Epidemiology and Biostatistics, University of California, San Francisco, CA
| | - Rulan S Parekh
- Hospital for Sick Children, University Healthy Network and University of Toronto, Toronto, Canada; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - W H Linda Kao
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Michael Bennett
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Mardge H Cohen
- Department of Medicine, Stroger Hospital, Rush University, Chicago, IL; Department of Medicine, Rush University, Chicago, IL
| | - Marek Nowicki
- Department of Medicine, University of Southern California, Los Angeles, CA
| | - Anjali Sharma
- Division of Infectious Diseases, Department of Medicine, SUNY Downstate Medical Center, Brooklyn, NY
| | - Mary Young
- Division of Infectious Diseases and Travel Medicine, Department of Medicine, Georgetown University Medical Center, Washington, DC
| | - Phyllis C Tien
- Department of Medicine, San Francisco VA Medical Center, San Francisco, CA
| | - Chirag R Parikh
- Section of Nephrology, Department of Medicine, Yale University, New Haven, CT; Program of Applied Translational Research, Yale University, New Haven, CT
| | | |
Collapse
|
42
|
Schiefner A, Skerra A. The menagerie of human lipocalins: a natural protein scaffold for molecular recognition of physiological compounds. Acc Chem Res 2015; 48:976-85. [PMID: 25756749 DOI: 10.1021/ar5003973] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
While immunoglobulins are well-known for their characteristic ability to bind macromolecular antigens (i.e., as antibodies during an immune response), the lipocalins constitute a family of proteins whose role is the complexation of small molecules for various physiological processes. In fact, a number of low-molecular-weight substances in multicellular organisms show poor solubility, are prone to chemical decomposition, or play a pathophysiological role and thus require specific binding proteins for transport through body fluids, storage, or sequestration. In many cases, lipocalins are involved in such tasks. Lipocalins are small, usually monomeric proteins with 150-180 residues and diameters of approximately 40 Å, adopting a compact fold that is dominated by a central eight-stranded up-and-down β-barrel. At the amino-terminal end, this core is flanked by a coiled polypeptide segment, while its carboxy-terminal end is followed by an α-helix that leans against the β-barrel as well as an amino acid stretch in a more-or-less extended conformation, which finally is fixed by a disulfide bond. Within the β-barrel, the antiparallel strands (designated A to H) are arranged in a (+1)7 topology and wind around a central axis in a right-handed manner such that part of strand A is hydrogen-bonded to strand H again. Whereas the lower region of the β-barrel is closed by short loops and densely packed hydrophobic side chains, including many aromatic residues, the upper end is usually open to solvent. There, four long loops, each connecting one pair of β-strands, together form the entrance to a cup-shaped cavity. Depending on the individual structure of a lipocalin, and especially on the lengths and amino acid sequences of its four loops, this pocket can accommodate chemical ligands of various sizes and shapes, including lipids, steroids, and other chemical hormones as well as secondary metabolites such as vitamins, cofactors, or odorants. While lipocalins are ubiquitous in all higher organisms, physiologically important members of this family have long been known in the human body, for example with the plasma retinol-binding protein that serves for the transport of vitamin A. This prototypic human lipocalin was the first for which a crystal structure was solved. Notably, several other lipocalins were discovered and assigned to this protein class before the term itself became familiar, which explains their diverse names in the scientific literature. To date, up to 15 distinct members of the lipocalin family have been characterized in humans, and during the last two decades the three-dimensional structures of a dozen major subtypes have been elucidated. This Account presents a comprehensive overview of the human lipocalins, revealing common structural principles but also deviations that explain individual functional features. Taking advantage of modern methods for combinatorial protein design, lipocalins have also been employed as scaffolds for the construction of artifical binding proteins with novel ligand specificities, so-called Anticalins, hence opening perspectives as a new class of biopharmaceuticals for medical therapy.
Collapse
Affiliation(s)
- André Schiefner
- Munich Center for Integrated
Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie, Technische Universität München, 85350 Freising-Weihenstephan, Germany
| | - Arne Skerra
- Munich Center for Integrated
Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie, Technische Universität München, 85350 Freising-Weihenstephan, Germany
| |
Collapse
|
43
|
Chiou SH, Huang CH, Liang SS. From Chemistry to Translational Medicine: The Application of Proteomics to Cancer Biomarker Discovery and Diagnosis. J CHIN CHEM SOC-TAIP 2015. [DOI: 10.1002/jccs.201400350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
44
|
Luczak M, Formanowicz D, Marczak Ł, Pawliczak E, Wanic-Kossowska M, Figlerowicz M, Stobiecki M. Deeper insight into chronic kidney disease-related atherosclerosis: comparative proteomic studies of blood plasma using 2DE and mass spectrometry. J Transl Med 2015; 13:20. [PMID: 25622820 PMCID: PMC4316657 DOI: 10.1186/s12967-014-0378-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 12/30/2014] [Indexed: 12/17/2022] Open
Abstract
Background Atherosclerosis is a major cause of cardiac events and mortality in patients suffering from chronic kidney disease (CKD). Moreover, the risk of cardiovascular disease (CVD) development in patients with CKD increases as kidney function declines. Although the close connection between atherosclerosis and kidney dysfunction is undeniable, particular risk factors and specific mechanisms that promote CVD in patients with CKD remain unclear. To gain insight into better recognition of the mechanisms of accelerated atherosclerosis in patients with CKD, we performed a comparative proteomic analysis of blood plasma from patients in various stages of CKD and thus distinct progression of atherosclerosis (n = 90), patients with advanced CVD and normal renal function (n = 30) and healthy volunteers (n = 30). Methods Plasma samples were depleted using affinity chromatography and divided into three fractions: high-abundant, low-abundant and low-molecular weight proteins. The first two fractions were analyzed by two-dimensional gel electrophoresis and mass spectrometry, the last one has been subjected to direct MS/MS analysis. A proteomic profiles for high-abundant, low-abundant and low-molecular weight proteins fractions were obtained. Differential accumulated proteins were confirmed by selected reaction monitoring analysis (SRM). The Gene Ontology (GO) function and the interaction networks of differentially expressed proteins were then analyzed. Results Forty-nine proteins (13 high- and 36 low-molecular mass) showed differences in accumulation levels. For eleven of them differential expression were confirmed by selected reaction monitoring analysis. Bioinformatic analysis showed that identified differential proteins were related to three different processes: the blood coagulation cascade, the transport, binding and metabolism of lipoproteins and inflammatory processes. Conclusions Obtained data provide an additional line of evidence that different molecular mechanisms are involved in the development of CKD- and CVD-related atherosclerosis. The abundance of some anti-atherogenic factors revealed in patients with CKD suggests that these factors are not associated with the reduction of atherosclerosis progression in CKD that is typically observed in “classical” CVD. Moreover, obtained data also suggest that mechanism of CVD acceleration may be different in initial and advanced stages of CKD. Undoubtedly, in advanced stages of CKD inflammation is highly pronounced. Electronic supplementary material The online version of this article (doi:10.1186/s12967-014-0378-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Magdalena Luczak
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704, Poznan, Poland. .,Institute of Chemical Technology and Engineering, Poznan University of Technology, Piotrowo 3A, 60-965, Poznan, Poland.
| | - Dorota Formanowicz
- Department of Clinical Biochemistry, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780, Poznan, Poland.
| | - Łukasz Marczak
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704, Poznan, Poland.
| | - Elżbieta Pawliczak
- Department of Nephrology, Transplantology and Internal Medicine, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355, Poznan, Poland.
| | - Maria Wanic-Kossowska
- Department of Nephrology, Transplantology and Internal Medicine, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355, Poznan, Poland.
| | - Marek Figlerowicz
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704, Poznan, Poland. .,Institute of Computing Science, Poznan University of Technology, Piotrowo 2, 60-965, Poznan, Poland.
| | - Maciej Stobiecki
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704, Poznan, Poland.
| |
Collapse
|
45
|
Jotwani V, Scherzer R, Abraham A, Estrella MM, Bennett M, Cohen MH, Nowicki M, Sharma A, Young M, Tien PC, Ix JH, Sarnak MJ, Parikh CR, Shlipak MG. Association of urine α1-microglobulin with kidney function decline and mortality in HIV-infected women. Clin J Am Soc Nephrol 2014; 10:63-73. [PMID: 25370597 DOI: 10.2215/cjn.03220314] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND OBJECTIVES Despite advances in therapy, HIV-infected individuals remain at higher risk for kidney dysfunction than uninfected individuals. It was hypothesized that urine levels of α1-microglobulin, a biomarker of proximal tubular dysfunction, would predict kidney function decline and mortality risk in HIV-infected and uninfected women. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS In the Women's Interagency HIV Study, urine α1-microglobulin and creatinine concentrations were measured in 903 HIV-infected and 287 uninfected women using stored urine from 1999 to 2000, when prevalence of tenofovir use was <1%. Participants were categorized into three categories by level of α1-microglobulin-to-creatinine ratio, and associations with kidney decline and all-cause mortality over 8 years were evaluated. RESULTS Urine α1-microglobulin was detectable in 60% of HIV-infected and 40% of uninfected women (P<0.001). Among HIV-infected women, there were 177 (22%), 61 (7%), and 128 (14%) patients with incident CKD, with 10% annual eGFR decline, and who died, respectively. Compared with HIV-infected women in the lowest α1-microglobulin category, HIV-infected women in the highest α1-microglobulin category had a 2.1-fold risk of incident CKD (95% confidence interval, 1.3 to 3.4), 2.7-fold risk of 10% annual eGFR decline (95% confidence interval, 1.2 to 5.9), and 1.6-fold mortality risk (95% confidence interval, 1.0 to 2.6) in models adjusting for kidney risk factors, baseline eGFR, and albuminuria. Among uninfected women, the highest α1-microglobulin category was associated with 3% (relative risk, 2.2; 95% confidence interval, 1.4 to 3.5) and 5% (relative risk, 2.2; 95% confidence interval, 1.1 to 4.3) annual eGFR decline relative to the lowest α1-microglobulin category. CONCLUSIONS Proximal tubular dysfunction, indicated by urine α1-microglobulin, was independently associated with kidney function decline in HIV-infected and uninfected women and mortality risk among HIV-infected women.
Collapse
Affiliation(s)
- Vasantha Jotwani
- Due to the number of contributing authors, the affiliations are provided in the Supplemental Material
| | - Rebecca Scherzer
- Due to the number of contributing authors, the affiliations are provided in the Supplemental Material
| | - Alison Abraham
- Due to the number of contributing authors, the affiliations are provided in the Supplemental Material
| | - Michelle M Estrella
- Due to the number of contributing authors, the affiliations are provided in the Supplemental Material
| | - Michael Bennett
- Due to the number of contributing authors, the affiliations are provided in the Supplemental Material
| | - Mardge H Cohen
- Due to the number of contributing authors, the affiliations are provided in the Supplemental Material
| | - Marek Nowicki
- Due to the number of contributing authors, the affiliations are provided in the Supplemental Material
| | - Anjali Sharma
- Due to the number of contributing authors, the affiliations are provided in the Supplemental Material
| | - Mary Young
- Due to the number of contributing authors, the affiliations are provided in the Supplemental Material
| | - Phyllis C Tien
- Due to the number of contributing authors, the affiliations are provided in the Supplemental Material
| | - Joachim H Ix
- Due to the number of contributing authors, the affiliations are provided in the Supplemental Material
| | - Mark J Sarnak
- Due to the number of contributing authors, the affiliations are provided in the Supplemental Material
| | - Chirag R Parikh
- Due to the number of contributing authors, the affiliations are provided in the Supplemental Material
| | - Michael G Shlipak
- Due to the number of contributing authors, the affiliations are provided in the Supplemental Material.
| |
Collapse
|
46
|
Huang H, Han Y, Gao J, Feng J, Zhu L, Qu L, Shen L, Shou C. High level of serum AMBP is associated with poor response to paclitaxel-capecitabine chemotherapy in advanced gastric cancer patients. Med Oncol 2013; 30:748. [PMID: 24135868 DOI: 10.1007/s12032-013-0748-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 10/02/2013] [Indexed: 12/13/2022]
Abstract
Gastric cancer is one of the most common human cancers and ranks the second in the global cancer-related mortality. The clinical outcome of patients with advanced gastric cancer (AGC) is markedly dependent on their response to the chemotherapy. Paclitaxel plus capecitabine, as a first-line regimen, is widely administrated in AGC patients, but more than a half of the patients have a poor response, possibly due to their resistance to the treatment. Therefore, it is important to identify potential responders to improve the efficacy of the chemotherapy. In the present study, we used an isobaric tag approach for relative and absolute quantification combined with ESI-QUAD-TOF/MS to identify potential predictive biomarkers for the chemotherapy. We found 211 serum proteins, and confirmed 17 candidates that were differentially present in the progression of disease (PD) group and the partial response (PR) group to the treatment of paclitaxel plus capecitabine. In further validation of the 17 candidates in the set of 12 PD and 12 PR AGC patients, we identified a higher level of AMBP (Alpha-1-Microglobulin/Bikunin Precursor) in the sera of PD patients than of the PR patients assayed by ELISA (9.13 ± 0.45 vs. 8.11 ± 0.26 μg/mL, p = 0.06) and by the Western blotting (relative gray value 396.4 ± 39.1 vs. 275.0 ± 34.76, p = 0.03), respectively. The receiver operating characteristics curve showed 75% sensitivity and 75% specificity of AMBP in AGC patients treated with the chemotherapy. Our data indicated that the high level of serum AMBP could predict the poor response of the AGC patients treated with the paclitaxel-capecitabine chemotherapy, which could be used as a potential biomarker to identify patients who would benefit from this chemotherapeutic regimen.
Collapse
Affiliation(s)
- Hao Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital and Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Use of α1-microglobulin for diagnosing chronic interstitial nephropathy. Clin Exp Med 2013; 14:315-20. [PMID: 23793995 DOI: 10.1007/s10238-013-0242-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Accepted: 05/28/2013] [Indexed: 10/26/2022]
Abstract
α1-Microglobulin (α1M) is a low molecular weight protein and has been best characterized for detecting acute lesions of proximal tubules (Bonventre in Contrib Nephrol 156:213-219, 2007). This study has tried to evaluate the use of α1M for the differential diagnosis of chronic interstitial nephropathy. 145 patients were recruited [81 men and 64 women, mean age 61.8 ± 16.7 years, 64.8 % have an estimated glomerular filtration (GFR) <60 ml/min]. Urinary α1M was evaluated using an immunonephelometric assay. 82 patients were diagnosed as having chronic interstitial nephritis (CIN), and 46 patients have been previously diagnosed of glomerulonephritis (GN). A group of hypertensive patients without renal disease was used as control (n = 17). Patients in GN group had the highest α1M excretion (15.05 mg/24 h). When the α1M/albuminuria rates were calculated, the CIN group had the highest rate (1.03 mg/mg) and the GN group had the lowest rate (0.04 mg/mg) (p < 0.001). When the α1M/proteinuria rates were calculated, the results were rather similar. The AUC for CIN group was 0.785, and the one for GN group was 0.139. Patients with estimated GFR <60 ml/min showed a higher excretion of α1M (18.75, 8.75-40.00 mg/24 h). Nevertheless, α1M/albuminuria and α1M/proteinuria rates were still higher in CIN patients with GFR ≥60 ml/min. α1M urinary excretion is increased in chronic interstitial nephropathy and glomerulonephritis as well as in patients with GFR <60 ml/min. The α1M/albuminuria rate and the α1M/proteinuria quotient are increased in chronic interstitial nephropathies but decreased in glomerular diseases.
Collapse
|
48
|
Weyer K, Nielsen R, Petersen SV, Christensen EI, Rehling M, Birn H. Renal uptake of 99mTc-dimercaptosuccinic acid is dependent on normal proximal tubule receptor-mediated endocytosis. J Nucl Med 2012; 54:159-65. [PMID: 23232279 DOI: 10.2967/jnumed.112.110528] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED (99m)Tc-labeled dimercaptosuccinic acid ((99m)Tc-DMSA) accumulates in the kidney cortex and is widely used for imaging of the renal parenchyma. Despite its extensive clinical use, the mechanism for renal targeting of the tracer is unresolved. Megalin and cubilin are cooperating receptors essential to the proximal tubule endocytic uptake of proteins from the glomerular ultrafiltrate. We have used megalin/cubilin-deficient mice produced by gene knockout to determine whether receptor-mediated endocytosis is responsible for the renal uptake of (99m)Tc-DMSA. METHODS Control or megalin/cubilin-deficient mice were injected intravenously with 0.5 MBq of (99m)Tc-DMSA or (99m)Tc-mercaptoacetyltriglycine (MAG3). Whole-body scintigrams and the activity in plasma, urine, and the kidneys were examined 6 h after injection. The size and identity of (99m)Tc-DMSA-bound proteins in urine were analyzed by fractionation by centrifugation and separation by sodium dodecyl sulfate polyacrylamide gel electrophoresis, followed by autoradiography and mass spectrometry. RESULTS No renal accumulation of (99m)Tc-DMSA was identified in scintigrams of megalin/cubilin-deficient mice. The renal accumulated activity of the tracer was reduced to 11.4% (± 2.5%, n = 7) of the normal uptake in control mice, correlating with a reduction in renal megalin/cubilin expression in knockout mice to about 10% of normal. The reduced renal uptake in megalin/cubilin-deficient mice was accompanied by an increase in the urinary excretion of (99m)Tc-DMSA. Size separation of the urine by ultracentrifugation and sodium dodecyl sulfate polyacrylamide gel electrophoresis demonstrated that in megalin/cubilin-deficient mice an increased amount of (99m)Tc-DMSA was excreted in an approximately 27-kDa form, which by mass spectrometry was identified as the plasma protein α1-microglobulin, an established megalin/cubilin ligand. CONCLUSION (99m)Tc-DMSA is filtered bound to α1-microglobulin and accumulates in the kidneys by megalin/cubilin-mediated endocytosis of the (99m)Tc-DMSA protein complex. Renal accumulation of (99m)Tc-DMSA is thus critically dependent on megalin/cubilin receptor function and therefore is a marker of proximal tubule endocytic activity.
Collapse
Affiliation(s)
- Kathrin Weyer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| | | | | | | | | | | |
Collapse
|
49
|
Nalepa AI, Taing JJ, Savitsky A, Knipp M. Preparation of cysteine-34-nitroxide spin labeled human α₁-microglobulin. Protein Expr Purif 2012. [PMID: 23201281 DOI: 10.1016/j.pep.2012.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
α(1)-Microglobulin (α(1)m) is a protein of yet unresolved function occurring in blood plasma and urine. It consists of a lipocaline type of fold with two cysteine residues forming a disulfide bridge and the third cysteine-34 remaining a free, somewhat reactive thiol. A number of investigations point to an interaction with heme and we have recently reported, that heme binding triggers the formation of a stable α(1)m trimer upon modification of cysteine-34 with 2-iodoacetamide, i.e., [α(1)m(heme)(2)](3) [J.F. Siebel, R.L. Kosinsky, B. Åkerström, M. Knipp, Insertion of heme b into the structure of the Cys34-carbamidomethylated human lipocalin α(1)-microglobulin-formation of a [(heme)(2)(α(1)-microglobulin)](3) complex, ChemBioChem 13 (2012) 879-887]. For further structural and functional investigations, an improved purification protocol for α(1)m was sought, in particular yielding an untagged amino acid sequence. The method reported herein improves the speed and the yield of the protein production even when an expression plasmid without tag was applied. Furthermore, for the purpose of future structural studies using electron paramagnetic resonance (EPR) techniques, in accordance to the modification with 2-iodoacetamide (α(1)m(AM)), the protein was modified with 3-(2-iodoacetamido)-2,2,5,5-tetramethyl-1-pyrrolidinyloxy (3-(2-iodoacetamido)-PROXYL) yielding the nitroxide spin labeled α(1)m(N-O). The extinction coefficient of the protein was calibrated using magnetic circular dichroism (MCD) spectroscopy of tryptophan (ε(280nm)=40,625M(-1)cm(-1)). The parallel quantification by absorbance spectroscopy (protein) and cw-EPR spectroscopy (radical spin) determined the degree of spin labeling to 90%. Characterization of the protein by circular dichroism (CD) spectroscopy and matrix assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) upon tryptic digestion further demonstrated the similar fold of α(1)m(AM) and α(1)m(N-O), but also established the modification of cystein-34 as well as the formation of the cysteine-72-cysteine-169 disulfide bond.
Collapse
Affiliation(s)
- Anna I Nalepa
- Max-Planck-Institut für Chemische Energiekonversion, Stiftstrasse 34-36, D-45470 Mülheim an der Ruhr, Germany
| | | | | | | |
Collapse
|
50
|
Siebel JF, Kosinsky RL, Åkerström B, Knipp M. Insertion of heme b into the structure of the Cys34-carbamidomethylated human lipocalin α(1)-microglobulin: formation of a [(heme)(2) (α(1)-Microglobulin)](3) complex. Chembiochem 2012; 13:879-87. [PMID: 22492620 DOI: 10.1002/cbic.201100808] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
α(1)-Microglobulin (α(1)m) is a 26 kDa plasma and tissue protein belonging to the lipocalin protein family. Previous investigations indicate that the protein interacts with heme and suggest that it has a function in heme metabolism. However, detailed characterizations of the α(1)m-heme interactions are lacking. Here, we report for the first time the preparation and analysis of a stable α(1)m-heme complex upon carbamidomethylation of the reactive Cys34 by using recombinantly expressed human α(1)m. Analytical size-exclusion chromatography coupled with a diode-array absorbance spectrophotometry demonstrates that at first an α(1)m-heme monomer is formed. Subsequently, a second heme triggers oligomerization that leads to trimerization. The resulting (α(1)m[heme](2))(3) complex was characterized by resonance Raman and EPR spectroscopy, which support the presence of two ferrihemes, thus indicating an unusual spin-state admixed ground state with S=(3)/(2), (5)/(2).
Collapse
Affiliation(s)
- Judith F Siebel
- Max-Planck-Institut für Bioanorganische Chemie, Stiftstrasse 34-36, 45470 Mülheim an der Ruhr, Germany
| | | | | | | |
Collapse
|