1
|
Pickkers P, Angus DC, Bass K, Bellomo R, van den Berg E, Bernholz J, Bestle MH, Doi K, Doig CJ, Ferrer R, Francois B, Gammelager H, Pedersen UG, Hoste E, Iversen S, Joannidis M, Kellum JA, Liu K, Meersch M, Mehta R, Millington S, Murray PT, Nichol A, Ostermann M, Pettilä V, Solling C, Winkel M, Young PJ, Zarbock A. Phase-3 trial of recombinant human alkaline phosphatase for patients with sepsis-associated acute kidney injury (REVIVAL). Intensive Care Med 2024; 50:68-78. [PMID: 38172296 PMCID: PMC10810941 DOI: 10.1007/s00134-023-07271-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/09/2023] [Indexed: 01/05/2024]
Abstract
PURPOSE Ilofotase alfa is a human recombinant alkaline phosphatase with reno-protective effects that showed improved survival and reduced Major Adverse Kidney Events by 90 days (MAKE90) in sepsis-associated acute kidney injury (SA-AKI) patients. REVIVAL, was a phase-3 trial conducted to confirm its efficacy and safety. METHODS In this international double-blinded randomized-controlled trial, SA-AKI patients were enrolled < 72 h on vasopressor and < 24 h of AKI. The primary endpoint was 28-day all-cause mortality. The main secondary endpoint was MAKE90, other secondary endpoints were (i) days alive and free of organ support through day 28, (ii) days alive and out of the intensive care unit (ICU) through day 28, and (iii) time to death through day 90. Prior to unblinding, the statistical analysis plan was amended, including an updated MAKE90 definition. RESULTS Six hundred fifty patients were treated and analyzed for safety; and 649 for efficacy data (ilofotase alfa n = 330; placebo n = 319). The observed mortality rates in the ilofotase alfa and placebo groups were 27.9% and 27.9% at 28 days, and 33.9% and 34.8% at 90 days. The trial was stopped for futility on the primary endpoint. The observed proportion of patients with MAKE90A and MAKE90B were 56.7% and 37.4% in the ilofotase alfa group vs. 64.6% and 42.8% in the placebo group. Median [interquartile range (IQR)] days alive and free of organ support were 17 [0-24] and 14 [0-24], number of days alive and discharged from the ICU through day 28 were 15 [0-22] and 10 [0-22] in the ilofotase alfa and placebo groups, respectively. Adverse events were reported in 67.9% and 75% patients in the ilofotase and placebo group. CONCLUSION Among critically ill patients with SA-AKI, ilofotase alfa did not improve day 28 survival. There may, however, be reduced MAKE90 events. No safety concerns were identified.
Collapse
Affiliation(s)
- Peter Pickkers
- Department of Intensive Care, Radboudumc, Nijmegen, The Netherlands.
| | - Derek C Angus
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Rinaldo Bellomo
- Department of Intensive Care, Austin Hospital, Melbourne, VIC, Australia
- Australian and New Zealand Intensive Care Research Centre, Monash University, Melbourne, VIC, Australia
| | | | | | - Morten H Bestle
- Department of Anesthesiology and Intensive Care, Copenhagen University Hospital-North Zealand, Hilleroed, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Kent Doi
- Department of Emergency and Critical Care Medicine, The University of Tokyo, Tokyo, Japan
| | - Chistopher J Doig
- Department of Critical Care Medicine, Medicine and Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Ricard Ferrer
- Department of Intensive Care Medicine, SODIR-VHIR Research Group, Val d'Hebron University Hospital, Barcelona, Spain
| | - Bruno Francois
- Intensive Care, Inserm CIC 1435 & UMR 1092, CHU Limoges, Limoges, France
| | - Henrik Gammelager
- Department of Intensive Care Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | - Eric Hoste
- Department of Internal Medicine and Pediatrics, Intensive Care Unit, Ghent University Hospital, Ghent University, Ghent, Belgium
- Research Foundation-Flanders, (FWO), Brussels, Belgium
| | - Susanne Iversen
- Department of Anaesthesiology and Intensive Care, Slagelse Hospital, Slagelse, Denmark
| | - Michael Joannidis
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - John A Kellum
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Kathleen Liu
- Division of Nephrology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Melanie Meersch
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Ravindra Mehta
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | | | | | - Alistair Nichol
- Australian and New Zealand Intensive Care Research Centre, Monash University, Melbourne, VIC, Australia
- University College Dublin-Clinical Research Centre at St Vincent's University Hospital, Dublin, Ireland
| | - Marlies Ostermann
- Department of Critical Care, Guys & St Thomas' Foundation Trust, London, UK
| | - Ville Pettilä
- Department of Perioperative and Intensive Care, University of Helsinki and Helsinki University Hospital, HUS, Helsinki, Finland
| | - Christoffer Solling
- Department of Anaestesiology and Intensive Care, Viborg Regional Hospital, Viborg, Denmark
| | | | - Paul J Young
- Australian and New Zealand Intensive Care Research Centre, Monash University, Melbourne, VIC, Australia
- Intensive Care Unit, Wellington Hospital, Wellington, New Zealand
- Medical Research Institute of New Zealand, Wellington, New Zealand
- Department of Critical Care, University of Melbourne, Melbourne, VIC, Australia
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| |
Collapse
|
2
|
Rosin DL, Hall JP, Zheng S, Huang L, Campos-Bilderback S, Sandoval R, Bree A, Beaumont K, Miller E, Larsen J, Hariri G, Kaila N, Encarnacion IM, Gale JD, van Elsas A, Molitoris BA, Okusa MD. Human Recombinant Alkaline Phosphatase (Ilofotase Alfa) Protects Against Kidney Ischemia-Reperfusion Injury in Mice and Rats Through Adenosine Receptors. Front Med (Lausanne) 2022; 9:931293. [PMID: 35966871 PMCID: PMC9366018 DOI: 10.3389/fmed.2022.931293] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/21/2022] [Indexed: 11/26/2022] Open
Abstract
Adenosine triphosphate (ATP) released from injured or dying cells is a potent pro-inflammatory "danger" signal. Alkaline phosphatase (AP), an endogenous enzyme that de-phosphorylates extracellular ATP, likely plays an anti-inflammatory role in immune responses. We hypothesized that ilofotase alfa, a human recombinant AP, protects kidneys from ischemia-reperfusion injury (IRI), a model of acute kidney injury (AKI), by metabolizing extracellular ATP to adenosine, which is known to activate adenosine receptors. Ilofotase alfa (iv) with or without ZM241,385 (sc), a selective adenosine A2A receptor (A2AR) antagonist, was administered 1 h before bilateral IRI in WT, A2AR KO (Adora2a-/- ) or CD73-/- mice. In additional studies recombinant alkaline phosphatase was given after IRI. In an AKI-on-chronic kidney disease (CKD) ischemic rat model, ilofotase alfa was given after the three instances of IRI and rats were followed for 56 days. Ilofotase alfa in a dose dependent manner decreased IRI in WT mice, an effect prevented by ZM241,385 and partially prevented in Adora2a-/- mice. Enzymatically inactive ilofotase alfa was not protective. Ilofotase alfa rescued CD73-/- mice, which lack a 5'-ectonucleotidase that dephosphorylates AMP to adenosine; ZM241,385 inhibited that protection. In both rats and mice ilofotase alfa ameliorated IRI when administered after injury, thus providing relevance for therapeutic dosing of ilofotase alfa following established AKI. In an AKI-on-CKD ischemic rat model, ilofotase alfa given after the third instance of IRI reduced injury. These results suggest that ilofotase alfa promotes production of adenosine from liberated ATP in injured kidney tissue, thereby amplifying endogenous mechanisms that can reverse tissue injury, in part through A2AR-and non-A2AR-dependent signaling pathways.
Collapse
Affiliation(s)
- Diane L. Rosin
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States,*Correspondence: Diane L. Rosin, , orcid.org/0000-0003-0187-5717
| | - J. Perry Hall
- Inflammation and Immunology Research Unit, Pfizer Inc., Cambridge, MA, United States
| | - Shuqiu Zheng
- Division of Nephrology, Center for Immunity, Inflammation and Regeneration, University of Virginia, Charlottesville, VA, United States
| | - Liping Huang
- Division of Nephrology, Center for Immunity, Inflammation and Regeneration, University of Virginia, Charlottesville, VA, United States
| | - Silvia Campos-Bilderback
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indiana Center for Biological Microscopy, Roudebush VA Medical Center, Indianapolis, IN, United States
| | - Ruben Sandoval
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indiana Center for Biological Microscopy, Roudebush VA Medical Center, Indianapolis, IN, United States
| | - Andrea Bree
- Inflammation and Immunology Research Unit, Pfizer Inc., Cambridge, MA, United States
| | - Kevin Beaumont
- BioMedicine Design, Pfizer Inc., Cambridge, MA, United States
| | - Emily Miller
- BioMedicine Design, Pfizer Inc., Groton, CT, United States
| | - Jennifer Larsen
- Early Clinical Development, Pfizer Inc., Groton, CT, United States
| | - Ghazal Hariri
- Drug Product Development, Pfizer Inc., Cambridge, MA, United States
| | - Neelu Kaila
- Medicinal Chemistry, Pfizer Inc., Cambridge, MA, United States
| | - Iain M. Encarnacion
- Division of Nephrology, Center for Immunity, Inflammation and Regeneration, University of Virginia, Charlottesville, VA, United States
| | - Jeremy D. Gale
- Inflammation and Immunology Research Unit, Pfizer Inc., Cambridge, MA, United States
| | | | - Bruce A. Molitoris
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indiana Center for Biological Microscopy, Roudebush VA Medical Center, Indianapolis, IN, United States
| | - Mark D. Okusa
- Division of Nephrology, Center for Immunity, Inflammation and Regeneration, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
3
|
Wu H, Wang Y, Li H, Meng L, Zheng N, Wang J. Protective Effect of Alkaline Phosphatase Supplementation on Infant Health. Foods 2022; 11:foods11091212. [PMID: 35563935 PMCID: PMC9101100 DOI: 10.3390/foods11091212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/13/2022] [Accepted: 04/13/2022] [Indexed: 12/03/2022] Open
Abstract
Alkaline phosphatase (ALP) is abundant in raw milk. Because of its high heat resistance, ALP negative is used as an indicator of successful sterilization. However, pasteurized milk loses its immune protection against allergy. Clinically, ALP is also used as an indicator of organ diseases. When the activity of ALP in blood increases, it is considered that diseases occur in viscera and organs. Oral administration or injecting ALP will not cause harm to the body and has a variety of probiotic effects. For infants with low immunity, ALP intake is a good prebiotic for protecting the infant’s intestine from potential pathogenic bacteria. In addition, ALP has a variety of probiotic effects for any age group, including prevention and treatment intestinal diseases, allergies, hepatitis, acute kidney injury (AKI), diabetes, and even the prevention of aging. The prebiotic effects of alkaline phosphatase on the health of infants and consumers and the content of ALP in different mammalian raw milk are summarized. The review calls on consumers and manufacturers to pay more attention to ALP, especially for infants with incomplete immune development. ALP supplementation is conducive to the healthy growth of infants.
Collapse
Affiliation(s)
- Haoming Wu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yang Wang
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China;
| | - Huiying Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lu Meng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nan Zheng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Correspondence: ; Tel.: +86-10-62816069
| | - Jiaqi Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
4
|
Abstract
INTRODUCTION Acute kidney injury (AKI) is a clinically critical disease exhibiting an acute decline in renal function. The lack of an effective prevention and treatment method equates to a high morbidity and mortality rate. Consequently, over the past few decades, many therapeutic drugs with different mechanisms of action have been proposed and gradually applied to the clinic. The involved drug mechanisms evaluated have included hemodynamic modulation, anti-inflammatory, antioxidant, repair agents, metabolic derangement and mitochondrial function. AREAS COVERED The authors of this review provide the reader with a reference point for the latest advances in pharmacotherapy in acute kidney injury. This is achieved by the evaluation of the latest data collected on potential therapeutic drugs with different mechanisms of action, as well as their preclinical and clinical impact on AKI. EXPERT OPINION Presently, the vast majority of drugs are still in clinical development, which is a huge challenge. Nevertheless, in addition to current chemical drugs and gene therapy strategies, the advent of mesenchymal stem cell treatments and other emerging pharmaceutical strategies could enable clinicians to better treat AKI. Due to the nonselective distribution and low bioavailability of some of the latest pharmaceutical strategies, there is hope that these treatment options may provide more efficacious avenues.
Collapse
Affiliation(s)
- Yali Xu
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ping Zou
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaojing Cao
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
5
|
Kühn F, Duan R, Ilmer M, Wirth U, Adiliaghdam F, Schiergens TS, Andrassy J, Bazhin AV, Werner J. Targeting the Intestinal Barrier to Prevent Gut-Derived Inflammation and Disease: A Role for Intestinal Alkaline Phosphatase. Visc Med 2021; 37:383-393. [PMID: 34722721 DOI: 10.1159/000515910] [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: 12/15/2020] [Accepted: 03/16/2021] [Indexed: 02/02/2023] Open
Abstract
Background Intestinal alkaline phosphatase (IAP) as a tissue-specific isozyme of alkaline phosphatases is predominantly produced by enterocytes in the proximal small intestine. In recent years, an increasing number of pathologies have been identified to be associated with an IAP deficiency, making it very worthwhile to review the various roles, biological functions, and potential therapeutic aspects of IAP. Summary IAP primarily originates and acts in the intestinal tract but affects other organs through specific biological axes related to its fundamental roles such as promoting gut barrier function, dephosphorylation/detoxification of lipopolysaccharides (LPS), and regulation of gut microbiota. Key Messages Numerous studies reporting on the different roles and the potential therapeutic value of IAP across species have been published during the last decade. While IAP deficiency is linked to varying degrees of physiological dysfunctions across multiple organ systems, the supplementation of IAP has been proven to be beneficial in several translational and clinical studies. The increasing evidence of the salutary functions of IAP underlines the significance of the naturally occurring brush border enzyme.
Collapse
Affiliation(s)
- Florian Kühn
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Ruifeng Duan
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Matthias Ilmer
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Ulrich Wirth
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Fatemeh Adiliaghdam
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tobias S Schiergens
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Joachim Andrassy
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Alexandr V Bazhin
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| | - Jens Werner
- Department of General, Visceral and Transplant Surgery, University Hospital of LMU Munich, Munich, Germany
| |
Collapse
|
6
|
Kidney Microcirculation as a Target for Innovative Therapies in AKI. J Clin Med 2021; 10:jcm10184041. [PMID: 34575154 PMCID: PMC8471583 DOI: 10.3390/jcm10184041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/29/2021] [Accepted: 09/02/2021] [Indexed: 12/19/2022] Open
Abstract
Acute kidney injury (AKI) is a serious multifactorial conditions accompanied by the loss of function and damage. The renal microcirculation plays a crucial role in maintaining the kidney’s functional and structural integrity for oxygen and nutrient supply and waste product removal. However, alterations in microcirculation and oxygenation due to renal perfusion defects, hypoxia, renal tubular, and endothelial damage can result in AKI and the loss of renal function regardless of systemic hemodynamic changes. The unique structural organization of the renal microvasculature and the presence of autoregulation make it difficult to understand the mechanisms and the occurrence of AKI following disorders such as septic, hemorrhagic, or cardiogenic shock; ischemia/reperfusion; chronic heart failure; cardiorenal syndrome; and hemodilution. In this review, we describe the organization of microcirculation, autoregulation, and pathophysiological alterations leading to AKI. We then suggest innovative therapies focused on the protection of the renal microcirculation and oxygenation to prevent AKI.
Collapse
|
7
|
Zhai M, Han M, Huang X, Kang F, Yang CH, Li J. Dexmedetomidine Protects Human Renal Tubular Epithelial HK-2 Cells against Hypoxia/Reoxygenation Injury by Inactivating Endoplasmic Reticulum Stress Pathway. CELL JOURNAL 2021; 23:457-464. [PMID: 34455722 PMCID: PMC8405080 DOI: 10.22074/cellj.2021.7220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 03/04/2020] [Indexed: 11/20/2022]
Abstract
Objective The study was aimed to investigate the effects and potential mechanisms of Dexmedetomidine (Dex) on
hypoxia/reoxygenation (H/R) injury in human renal tubular epithelial HK-2 cells. Materials and Methods In this experimental study, HK-2 cells were divided into four groups: control group, Dex
group, H/R group, and Dex+H/R group. The cells in control group received no treatment, and cells in Dex group were
only treated with 0.1 nmol/L Dex. The cells in H/R group and Dex+H/R group were all treated with H/R (hypoxia for
24 hours and normoxia for 4 hours), and only the cells in Dex+H/R group were pre-administrated with 0.1 nmol/L
Dex. Following treatments at 37˚C for 28 hours, cell viability and apoptosis were measured by MTT assay and flow
cytometry, respectively. Also, the expressions of hypoxia-inducible factor 1 (HIF-1α), glucose-regulated protein 78
(GRP78), C/EBP homologous protein (CHOP), caspase-12 and cleaved caspase-3 were determined by western blot.
Results The cell viability was significant decreased in H/R group compared with control group (P<0.05), while was
significantly increased in Dex+H/R group compared with that in H/R group (P<0.05). However, the change tendency
of the cell apoptosis was opposite to that of cell viability. Compared with H/R group, the expression of HIF-1α was
evidently up-regulated, while GRP78, CHOP, capase-12 and cleaved caspase-3 expressions were all obviously down-
regulated in Dex+H/R group (P<0.05). In addition, the concentrations of malondialdehyde (MDA) in H/R group and
Dex+H/R group were 1.68 ± 0.22 nmol/mgprot and 0.85 ± 0.16 nmol/mgprot, respectively. The superoxide dismutase
(SOD) activity was higher in Dex+H/R group (121 ± 11 U/L), which which was more than twice larger than that in H/R
group (57 ± 10 U/L).
Conclusion Dex could promote cell viability and inhibit apoptosis through up-regulating HIF-1α, reducing endoplasmic
reticulum (ER) stress and mediating oxidative stress, thus ameliorating the H/R injury.
Collapse
Affiliation(s)
- Mingyu Zhai
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Mingming Han
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiang Huang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Fang Kang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - C Hengwei Yang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Juan Li
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
8
|
Ren L, Ye J. The progress of research into the use of alkaline phosphatase in the treatment of sepsis-associated acute kidney injury. Asian J Surg 2021; 44:1212-1213. [PMID: 34210548 DOI: 10.1016/j.asjsur.2021.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 10/21/2022] Open
Affiliation(s)
- Liping Ren
- Dalian Medical University, Dalian, 116044, China
| | - Jilu Ye
- Department of Critical Care Medicine, Taizhou People's Hospital, Taizhou, 225300, China.
| |
Collapse
|
9
|
Nwafor DC, Brichacek AL, Ali A, Brown CM. Tissue-Nonspecific Alkaline Phosphatase in Central Nervous System Health and Disease: A Focus on Brain Microvascular Endothelial Cells. Int J Mol Sci 2021; 22:5257. [PMID: 34067629 PMCID: PMC8156423 DOI: 10.3390/ijms22105257] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/21/2022] Open
Abstract
Tissue-nonspecific alkaline phosphatase (TNAP) is an ectoenzyme bound to the plasma membranes of numerous cells via a glycosylphosphatidylinositol (GPI) moiety. TNAP's function is well-recognized from earlier studies establishing its important role in bone mineralization. TNAP is also highly expressed in cerebral microvessels; however, its function in brain cerebral microvessels is poorly understood. In recent years, few studies have begun to delineate a role for TNAP in brain microvascular endothelial cells (BMECs)-a key component of cerebral microvessels. This review summarizes important information on the role of BMEC TNAP, and its implication in health and disease. Furthermore, we discuss current models and tools that may assist researchers in elucidating the function of TNAP in BMECs.
Collapse
Affiliation(s)
- Divine C. Nwafor
- Department of Neuroscience, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA; (D.C.N.); (A.A.)
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Allison L. Brichacek
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA;
| | - Ahsan Ali
- Department of Neuroscience, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA; (D.C.N.); (A.A.)
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Candice M. Brown
- Department of Neuroscience, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA; (D.C.N.); (A.A.)
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA;
| |
Collapse
|
10
|
Juschten J, Ingelse SA, Bos LDJ, Girbes ARJ, Juffermans NP, van der Poll T, Schultz MJ, Tuinman PR. Alkaline phosphatase in pulmonary inflammation-a translational study in ventilated critically ill patients and rats. Intensive Care Med Exp 2020; 8:46. [PMID: 33336319 PMCID: PMC7746537 DOI: 10.1186/s40635-020-00335-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/31/2022] Open
Abstract
Background Alkaline phosphatase (AP), a dephosphorylating enzyme, is involved in various physiological processes and has been shown to have anti-inflammatory effects. Aim To determine the correlation between pulmonary AP activity and markers of inflammation in invasively ventilated critically ill patients with or without acute respiratory distress syndrome (ARDS), and to investigate the effect of administration of recombinant AP on pulmonary inflammation in a well-established lung injury model in rats Methods AP activity was determined and compared with levels of various inflammatory mediators in bronchoalveolar lavage fluid (BALF) samples obtained from critically ill patients within 2 days of start of invasive ventilation. The endpoints of this part of the study were the correlations between AP activity and markers of inflammation, i.e., interleukin (IL)-6 levels in BALF. In RccHan Wistar rats, lung injury was induced by intravenous administration of 10 mg/kg lipopolysaccharide, followed by ventilation with a high tidal volume for 4 h. Rats received either an intravenous bolus of 1500 IU/kg recombinant AP or normal saline 2 h after intravenous LPS administration, right before start of ventilation. Endpoints of this part of the study were pulmonary levels of markers of inflammation, including IL-6, and markers of endothelial and epithelial dysfunction. Results BALF was collected from 83 patients; 10 patients had mild ARDS, and 15 had moderate to severe ARDS. AP activity correlated well with levels of IL-6 (r = 0.70), as well as with levels of other inflammatory mediators. Pulmonary AP activity between patients with and without ARDS was comparable (0.33 [0.14–1.20] vs 0.55 [0.21–1.42] U/L; p = 0.37). Animals with acute lung injury had markedly elevated pulmonary AP activity compared to healthy controls (2.58 [2.18–3.59] vs 1.01 [0.80–1.46] U/L; p < 0.01). Intravenous administration of recombinant AP did neither affect pulmonary inflammation nor endothelial and epithelial dysfunction. Conclusions In ventilated critically ill patients, pulmonary AP activity correlates well with markers of pulmonary inflammation, such as IL-6 and IL-8. In animals with lung injury, pulmonary AP activity is elevated. Administration of recombinant AP does not alter pulmonary inflammation and endothelial or epithelial dysfunction in the acute phase of a murine lung injury model.
Collapse
Affiliation(s)
- Jenny Juschten
- Department of Intensive Care, Amsterdam University Medical Centers, location "VU", Mail stop ZH 7D-172, De Boelelaan 1117, 1082 RW, Amsterdam, the Netherlands. .,Research VUmc Intensive Care (REVIVE), Amsterdam University Medical Centers, location "VU", Amsterdam, the Netherlands. .,Department of Intensive Care, Amsterdam University Medical Centers, location "AMC", Amsterdam, the Netherlands. .,Laboratory of Experimental Intensive Care and Anesthesiology (L⋅E⋅I⋅C⋅A), Amsterdam University Medical Centers, location "AMC", Amsterdam, the Netherlands.
| | - Sarah A Ingelse
- Laboratory of Experimental Intensive Care and Anesthesiology (L⋅E⋅I⋅C⋅A), Amsterdam University Medical Centers, location "AMC", Amsterdam, the Netherlands.,Emma Children's Hospital-Pediatric Intensive Care Unit, Amsterdam University Medical Centers, location "AMC", Amsterdam, the Netherlands
| | - Lieuwe D J Bos
- Department of Pulmonology, Amsterdam University Medical Centers, location "AMC", Amsterdam, the Netherlands
| | - Armand R J Girbes
- Department of Intensive Care, Amsterdam University Medical Centers, location "VU", Mail stop ZH 7D-172, De Boelelaan 1117, 1082 RW, Amsterdam, the Netherlands.,Research VUmc Intensive Care (REVIVE), Amsterdam University Medical Centers, location "VU", Amsterdam, the Netherlands
| | - Nicole P Juffermans
- Laboratory of Experimental Intensive Care and Anesthesiology (L⋅E⋅I⋅C⋅A), Amsterdam University Medical Centers, location "AMC", Amsterdam, the Netherlands.,Department of Intensive Care, OLVG hospital, Amsterdam, The Netherlands
| | - Tom van der Poll
- Division of Infectious Diseases, Amsterdam University Medical Centers, location "AMC", Amsterdam, The Netherlands.,Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers, location "AMC", Amsterdam, The Netherlands
| | - Marcus J Schultz
- Department of Intensive Care, Amsterdam University Medical Centers, location "AMC", Amsterdam, the Netherlands.,Laboratory of Experimental Intensive Care and Anesthesiology (L⋅E⋅I⋅C⋅A), Amsterdam University Medical Centers, location "AMC", Amsterdam, the Netherlands.,Mahidol-Oxford Tropical Medicine Research Unit (MORU), Mahidol University, Bangkok, Thailand.,Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Pieter Roel Tuinman
- Department of Intensive Care, Amsterdam University Medical Centers, location "VU", Mail stop ZH 7D-172, De Boelelaan 1117, 1082 RW, Amsterdam, the Netherlands.,Research VUmc Intensive Care (REVIVE), Amsterdam University Medical Centers, location "VU", Amsterdam, the Netherlands
| | | |
Collapse
|
11
|
Alvarenga L, Cardozo LFMF, Lindholm B, Stenvinkel P, Mafra D. Intestinal alkaline phosphatase modulation by food components: predictive, preventive, and personalized strategies for novel treatment options in chronic kidney disease. EPMA J 2020; 11:565-579. [PMID: 33240450 PMCID: PMC7680467 DOI: 10.1007/s13167-020-00228-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 10/30/2020] [Indexed: 12/18/2022]
Abstract
Alkaline phosphatase (AP) is a ubiquitous membrane-bound glycoprotein that catalyzes phosphate monoesters' hydrolysis from organic compounds, an essential process in cell signaling. Four AP isozymes have been described in humans, placental AP, germ cell AP, tissue nonspecific AP, and intestinal AP (IAP). IAP plays a crucial role in gut microbial homeostasis, nutrient uptake, and local and systemic inflammation, and its dysfunction is associated with persistent inflammatory disorders. AP is a strong predictor of mortality in the general population and patients with cardiovascular and chronic kidney disease (CKD). However, little is known about IAP modulation and its possible consequences in CKD, a disease characterized by gut microbiota imbalance and persistent low-grade inflammation. Mitigating inflammation and dysbiosis can prevent cardiovascular complications in patients with CKD, and monitoring factors such as IAP can be useful for predicting those complications. Here, we review IAP's role and the results of nutritional interventions targeting IAP in experimental models to prevent alterations in the gut microbiota, which could be a possible target of predictive, preventive, personalized medicine (PPPM) to avoid CKD complications. Microbiota and some nutrients may activate IAP, which seems to have a beneficial impact on health; however, data on CKD remains scarce.
Collapse
Affiliation(s)
- L. Alvarenga
- Post Graduation Program in Medical Sciences, (UFF) Federal Fluminense University Niterói-Rio de Janeiro (RJ), Niterói, Brazil
| | - L. F. M. F. Cardozo
- Post Graduation Program in Cardiovascular Sciences, Federal Fluminense University (UFF), Niterói, Rio de Janeiro (RJ) Brazil
| | - B. Lindholm
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Technology and Intervention, Karolinska Institutet, Stockholm, Sweden
| | - P. Stenvinkel
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Technology and Intervention, Karolinska Institutet, Stockholm, Sweden
| | - D. Mafra
- Post Graduation Program in Medical Sciences, (UFF) Federal Fluminense University Niterói-Rio de Janeiro (RJ), Niterói, Brazil
- Post Graduation Program in Cardiovascular Sciences, Federal Fluminense University (UFF), Niterói, Rio de Janeiro (RJ) Brazil
| |
Collapse
|
12
|
Asad A, Burton JO, March DS. Exercise as a therapeutic option for acute kidney injury: mechanisms and considerations for the design of future clinical studies. BMC Nephrol 2020; 21:446. [PMID: 33097033 PMCID: PMC7585193 DOI: 10.1186/s12882-020-02098-9] [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: 05/11/2020] [Accepted: 10/09/2020] [Indexed: 12/18/2022] Open
Abstract
Acute kidney injury (AKI) is a known risk factor for chronic kidney disease (CKD) and end stage kidney disease (ESKD). The progression from AKI to CKD, despite being well recognised, is not completely understood, although sustained inflammation and fibrosis are implicated. A therapeutic intervention targeting the post AKI stage could reduce the progression to CKD, which has high levels of associated morbidity and mortality. Exercise has known anti-inflammatory effects with animal AKI models demonstrating its use as a therapeutic agent in abrogating renal injury. This suggests the use of an exercise rehabilitation programme in AKI patients following discharge could attenuate renal damage and improve long term patient outcomes. In this review article we outline considerations for future clinical studies of exercise in the AKI population.
Collapse
Affiliation(s)
- Anam Asad
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - James O Burton
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK.,NIHR Leicester Biomedical Research Centre, Leicester, UK.,School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Daniel S March
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK. .,NIHR Leicester Biomedical Research Centre, Leicester, UK.
| |
Collapse
|
13
|
Khailova L, Robison J, Jaggers J, Ing R, Lawson S, Treece A, Soranno D, Osorio Lujan S, Davidson JA. Tissue alkaline phosphatase activity and expression in an experimental infant swine model of cardiopulmonary bypass with deep hypothermic circulatory arrest. JOURNAL OF INFLAMMATION-LONDON 2020; 17:27. [PMID: 32817746 PMCID: PMC7422466 DOI: 10.1186/s12950-020-00256-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/30/2020] [Indexed: 01/12/2023]
Abstract
Background Infant cardiac surgery with cardiopulmonary bypass results in decreased circulating alkaline phosphatase that is associated with poor postoperative outcomes. Bovine intestinal alkaline phosphatase infusion represents a novel therapy for post-cardiac surgery organ injury. However, the effects of cardiopulmonary bypass and bovine-intestinal alkaline phosphatase infusion on tissue-level alkaline phosphatase activity/expression are unknown. Methods Infant pigs (n = 20) underwent cardiopulmonary bypass with deep hypothermic circulatory arrest followed by four hours of intensive care. Seven control animals underwent mechanical ventilation only. Cardiopulmonary bypass/deep hypothermic circulatory arrest animals were given escalating doses of bovine intestinal alkaline phosphatase infusion (0-25 U/kg/hr.; n = 5/dose). Kidney, liver, ileum, jejunum, colon, heart and lung were collected for measurement of tissue alkaline phosphatase activity and mRNA. Results Tissue alkaline phosphatase activity varied significantly across organs with the highest levels found in the kidney and small intestine. Cardiopulmonary bypass with deep hypothermic circulatory arrest resulted in decreased kidney alkaline phosphatase activity and increased lung alkaline phosphatase activity, with no significant changes in the other organs. Alkaline phosphatase mRNA expression was increased in both the lung and the ileum. The highest dose of bovine intestinal alkaline phosphatase resulted in increased kidney and liver tissue alkaline phosphatase activity. Conclusions Changes in alkaline phosphatase activity after cardiopulmonary bypass with deep hypothermic circulatory arrest and bovine intestinal alkaline phosphatase delivery are tissue specific. Kidneys, lung, and ileal alkaline phosphatase appear most affected by cardiopulmonary bypass with deep hypothermic circulatory arrest and further research is warranted to determine the mechanism and biologic importance of these changes.
Collapse
Affiliation(s)
- Ludmila Khailova
- Department of Pediatrics, University of Colorado, 13123 East 16th Ave, Box 100, Aurora, CO 80045 USA
| | - Justin Robison
- Department of Pediatrics, University of Colorado, 13123 East 16th Ave, Box 100, Aurora, CO 80045 USA
| | - James Jaggers
- Department of Surgery, University of Colorado, Aurora, CO USA
| | - Richard Ing
- Department of Anesthesiology, University of Colorado, Aurora, CO USA
| | - Scott Lawson
- Children's Hospital Colorado, Heart Institute, Aurora, CO USA
| | - Amy Treece
- Department of Pathology, University of Colorado, Aurora, CO USA
| | - Danielle Soranno
- Department of Pediatrics, University of Colorado, 13123 East 16th Ave, Box 100, Aurora, CO 80045 USA
| | - Suzanne Osorio Lujan
- Department of Pediatrics, University of Colorado, 13123 East 16th Ave, Box 100, Aurora, CO 80045 USA
| | - Jesse A Davidson
- Department of Pediatrics, University of Colorado, 13123 East 16th Ave, Box 100, Aurora, CO 80045 USA
| |
Collapse
|
14
|
Recombinant Alkaline Phosphatase Prevents Acute on Chronic Liver Failure. Sci Rep 2020; 10:389. [PMID: 31942020 PMCID: PMC6962206 DOI: 10.1038/s41598-019-57284-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 11/09/2019] [Indexed: 12/11/2022] Open
Abstract
The lipopolysaccharide (LPS)– toll-like receptor-4 (TLR4) pathway plays an important role in liver failure. Recombinant alkaline phosphatase (recAP) deactivates LPS. The aim of this study was to determine whether recAP prevents the progression of acute and acute-on-chronic liver failure (ACLF). Eight groups of rats were studied 4-weeks after sham surgery or bile duct ligation and were injected with saline or LPS to mimic ACLF. Acute liver failure was induced with Galactosamine-LPS and in both models animals were treated with recAP prior to LPS administration. In the ACLF model, the severity of liver dysfunction and brain edema was attenuated by recAP, associated with reduction in cytokines, chemokines, liver cell death, and brain water. The activity of LPS was reduced by recAP. The treatment was not effective in acute liver failure. Hepatic TLR4 expression was reduced by recAP in ACLF but not acute liver failure. Increased sensitivity to endotoxins in cirrhosis is associated with upregulation of hepatic TLR4, which explains susceptibility to development of ACLF whereas acute liver failure is likely due to direct hepatoxicity. RecAP prevents multiple organ injury by reducing receptor expression and is a potential novel treatment option for prevention of ACLF but not acute liver failure.
Collapse
|
15
|
Alkaline Phosphatase Treatment of Acute Kidney Injury in an Infant Piglet Model of Cardiopulmonary Bypass with Deep Hypothermic Circulatory Arrest. Sci Rep 2019; 9:14175. [PMID: 31578351 PMCID: PMC6775126 DOI: 10.1038/s41598-019-50481-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 09/13/2019] [Indexed: 01/11/2023] Open
Abstract
Acute kidney injury (AKI) is associated with prolonged hospitalization and mortality following infant cardiac surgery, but therapeutic options are limited. Alkaline phosphatase (AP) infusion reduced AKI in phase 2 sepsis trials but has not been evaluated for cardiac surgery-induced AKI. We developed a porcine model of infant cardiopulmonary bypass (CPB) with deep hypothermic circulatory arrest (DHCA) to investigate post-CPB/DHCA AKI, measure serum/renal tissue AP activity with escalating doses of AP infusion, and provide preliminary assessment of AP infusion for prevention of AKI. Infant pigs underwent CPB with DHCA followed by survival for 4 h. Groups were treated with escalating doses of bovine intestinal AP (1, 5, or 25U/kg/hr). Anesthesia controls were mechanically ventilated for 7 h without CPB. CPB/DHCA animals demonstrated histologic and biomarker evidence of AKI as well as decreased serum and renal tissue AP compared to anesthesia controls. Only high dose AP infusion significantly increased serum or renal tissue AP activity. Preliminary efficacy evaluation demonstrated a trend towards decreased AKI in the high dose AP group. The results of this dose-finding study indicate that AP infusion at the dose of 25U/kg/hr corrects serum and tissue AP deficiency and may prevent AKI in this piglet model of infant CPB/DHCA.
Collapse
|
16
|
Hümmeke-Oppers F, Hemelaar P, Pickkers P. Innovative Drugs to Target Renal Inflammation in Sepsis: Alkaline Phosphatase. Front Pharmacol 2019; 10:919. [PMID: 31507417 PMCID: PMC6716471 DOI: 10.3389/fphar.2019.00919] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 07/22/2019] [Indexed: 12/20/2022] Open
Abstract
Sepsis-related mortality roughly doubles when acute kidney injury (AKI) occurs and end-stage renal disease is more common in sepsis-associated AKI survivors. So far, no licensed treatment for the prevention of AKI is available, however the data on alkaline phosphatase (AP) is promising and might change this. Sepsis-associated AKI is believed to be the result of inflammation and hypoxia combined. Systemic inflammation started by recognition of ‘pathogen-associated molecular patterns’ (PAMPs) such as lipopolysaccharide (LPS) which binds to Toll-like receptor 4 and leads to the production of inflammatory mediators. Due to this inflammatory process renal microcirculation gets impaired leading to hypoxia resulting in cell damage or cell death. In the process of cell damage so called ‘danger-associated molecular patterns’ (DAMPs) are released resulting in a sustained inflammatory effect. Apart from the systemic inflammation DAMPs and PAMPs also interact with receptors in the proximal tubule of the kidney causing a local inflammatory response leading to leukocyte infiltration and tubular lesions, combined with renal cell apoptosis and ultimately to AKI. In the longer-term, inflammation-mediated inadequate repair mechanism may lead to fibrosis and development of chronic kidney disease. AP is an endogenous enzyme that dephosphorylates and thereby detoxifies several compounds, including LPS. A small phase 2 clinical trial in sepsis patients showed that urinary excretion of tubular injury markers was attenuated and creatinine clearance improved in sepsis patients treated with AP. This renal protective effect was confirmed in a second small clinical phase 2 trial in sepsis patients with AKI. Subsequently, a large trial in sepsis patients with AKI was conducted using a human recombinant AP. In 301 patients no improvement of kidney function within 7 days after enrolment was observed, but kidney function was significantly better on day 21 and day 28 and all-cause 28-day mortality was significantly lower (14.4% in AP group versus 26.7% in the placebo group). Possible explanations of this lack of short-term kidney function improvement are discussed and potential effects of AP on renal repair mechanisms, including inflammation-mediated induction of fibrosis, that may explain the beneficial longer-term effects of AP are proposed.
Collapse
Affiliation(s)
- Femke Hümmeke-Oppers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Pleun Hemelaar
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
17
|
Xiang L, Thompson MS, Clemmer JS, Mittwede PN, Khan T, Hester RL. Early treatment with GLP-1 after severe trauma preserves renal function in obese Zucker rats. Am J Physiol Regul Integr Comp Physiol 2019; 316:R621-R627. [PMID: 30811247 DOI: 10.1152/ajpregu.00312.2018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Early posttrauma hyperglycemia (EPTH) is correlated with later adverse outcomes, including acute kidney injury (AKI). Controlling EPTH in the prehospital setting is difficult because of the variability in the ideal insulin dosage and the potential risk of hypoglycemia, especially in those with confounding medical comorbidities of obesity and insulin resistance. Glucagon-like peptide-1 (GLP-1) controls glucose levels in a glucose-dependent manner and is a current target in antidiabetic therapy. We have shown that after orthopedic trauma, obese Zucker rats exhibit EPTH and a later development of AKI (within 24 h). We hypothesized that GLP-1 treatment after trauma decreases EPTH and protects renal function in obese Zucker rats. Obese Zucker rats (~12 wk old) were fasted for 4 h before trauma. Soft tissue injury, fibula fracture, and homogenized bone component injection were then performed in both hind limbs to induce severe extremity trauma. Plasma glucose levels were measured before and 15, 30, 60, 120, 180, 240, and 300 min after trauma. GLP-1 (3 μg·kg-1·h-1, 1.5 ml/kg total) or saline was continuously infused from 30 min to 5 h after trauma. Afterwards, rats were placed in metabolic cages overnight for urine collection. The following day, plasma interleukin (IL)-6 levels, renal blood flow (RBF), glomerular filtration rate (GFR), and renal oxygen delivery (Do2) and consumption (V̇o2) were measured. EPTH was evident within 15 min after trauma but was significantly ameliorated during the 5 h of GLP-1 infusion. One day after trauma, plasma IL-6 was markedly increased in the trauma group and decreased in GLP-1-treated animals. RBF, GFR, and Do2 all significantly decreased with trauma, but renal V̇o2 was unchanged. GLP-1 treatment normalized RBF, GFR, and Do2 without affecting V̇o2. These results suggest that GLP-1 decreases EPTH and protects against a later development of AKI. Early treatment with GLP-1 (or its analogs) to rapidly, effectively, and safely control EPTH may be beneficial in the prehospital care of obese patients after trauma.
Collapse
Affiliation(s)
- Lusha Xiang
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi.,United States Army Institute of Surgical Research , San Antonio, Texas
| | - Michael S Thompson
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - John S Clemmer
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - Peter N Mittwede
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center , Pittsburgh, Pennsylvania
| | - Tazim Khan
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - Robert L Hester
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| |
Collapse
|
18
|
Zhang Z. Human recombinant alkaline phosphatase: a promising, yet-to-be-tested agent for the treatment sepsis-induced acute kidney injury. ANNALS OF TRANSLATIONAL MEDICINE 2019; 6:S124. [PMID: 30740445 DOI: 10.21037/atm.2018.12.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Zhongheng Zhang
- Department of emergency medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| |
Collapse
|
19
|
Brichacek AL, Brown CM. Alkaline phosphatase: a potential biomarker for stroke and implications for treatment. Metab Brain Dis 2019; 34:3-19. [PMID: 30284677 PMCID: PMC6351214 DOI: 10.1007/s11011-018-0322-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 09/24/2018] [Indexed: 12/14/2022]
Abstract
Stroke is the fifth leading cause of death in the U.S., with more than 100,000 deaths annually. There are a multitude of risks associated with stroke, including aging, cardiovascular disease, hypertension, Alzheimer's disease (AD), and immune suppression. One of the many challenges, which has so far proven to be unsuccessful, is the identification of a cost-effective diagnostic or prognostic biomarker for stroke. Alkaline phosphatase (AP), an enzyme first discovered in the 1920s, has been evaluated as a potential biomarker in many disorders, including many of the co-morbidities associated with stroke. This review will examine the basic biology of AP, and its most common isoenzyme, tissue nonspecific alkaline phosphatase (TNAP), with a specific focus on the central nervous system. It examines the preclinical and clinical evidence which supports a potential role for AP in stroke and suggests potential mechanism(s) of action for AP isoenzymes in stroke. Lastly, the review speculates on the clinical utility of AP isoenzymes as potential blood biomarkers for stroke or as AP-targeted treatments for stroke patients.
Collapse
Affiliation(s)
- Allison L Brichacek
- Department of Microbiology, Immunology, and Cell Biology, Center for Basic and Translational Stroke Research, WVU Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Box 9177, Morgantown, WV, 26506, USA
- Department of Neuroscience, Emergency Medicine, and Microbiology, Immunology and Cell Biology, Center for Basic and Translational Stroke Research, WVU Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Box 9303, Morgantown, WV, 26506, USA
| | - Candice M Brown
- Department of Microbiology, Immunology, and Cell Biology, Center for Basic and Translational Stroke Research, WVU Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Box 9177, Morgantown, WV, 26506, USA.
- Department of Neuroscience, Emergency Medicine, and Microbiology, Immunology and Cell Biology, Center for Basic and Translational Stroke Research, WVU Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Box 9303, Morgantown, WV, 26506, USA.
| |
Collapse
|
20
|
Michel T, Joannes-Boyau O, Schneider AG. A cure for septic AKI: Why not keep the dream alive? Anaesth Crit Care Pain Med 2019; 38:1-2. [PMID: 30635097 DOI: 10.1016/j.accpm.2018.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Thibault Michel
- Service de médecine intensive adulte, CHU Vaudois (CHUV), Lausanne , Switzerland
| | - Olivier Joannes-Boyau
- CHU de Bordeaux, Service d'Anesthesie-Reanimation SUD, Hôpital Magellan, 33000 Bordeaux, France.
| | | |
Collapse
|
21
|
Bowden SA, Foster BL. Alkaline Phosphatase Replacement Therapy for Hypophosphatasia in Development and Practice. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1148:279-322. [PMID: 31482504 DOI: 10.1007/978-981-13-7709-9_13] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Hypophosphatasia (HPP) is an inherited disorder that affects bone and tooth mineralization characterized by low serum alkaline phosphatase. HPP is caused by loss-of-function mutations in the ALPL gene encoding the protein, tissue-nonspecific alkaline phosphatase (TNSALP). TNSALP is expressed by mineralizing cells of the skeleton and dentition and is associated with the mineralization process. Generalized reduction of activity of the TNSALP leads to accumulation of its substrates, including inorganic pyrophosphate (PPi) that inhibits physiological mineralization. This leads to defective skeletal mineralization, with manifestations including rickets, osteomalacia, fractures, and bone pain, all of which can result in multi-systemic complications with significant morbidity, as well as mortality in severe cases. Dental manifestations are nearly universal among affected individuals and feature most prominently premature loss of deciduous teeth. Management of HPP has been limited to supportive care until the introduction of a TNSALP enzyme replacement therapy (ERT), asfotase alfa (AA). AA ERT has proven to be transformative, improving survival in severely affected infants and increasing overall quality of life in children and adults with HPP. This chapter provides an overview of TNSALP expression and functions, summarizes HPP clinical types and pathologies, discusses early attempts at therapies for HPP, summarizes development of HPP mouse models, reviews design and validation of AA ERT, and provides up-to-date accounts of AA ERT efficacy in clinical trials and case reports, including therapeutic response, adverse effects, limitations, and potential future directions in therapy.
Collapse
Affiliation(s)
- S A Bowden
- Division of Endocrinology, Department of Pediatrics, Nationwide Children's Hospital/The Ohio State University College of Medicine, Columbus, OH, USA.
| | - B L Foster
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
22
|
Pickkers P, Mehta RL, Murray PT, Joannidis M, Molitoris BA, Kellum JA, Bachler M, Hoste EAJ, Hoiting O, Krell K, Ostermann M, Rozendaal W, Valkonen M, Brealey D, Beishuizen A, Meziani F, Murugan R, de Geus H, Payen D, van den Berg E, Arend J. Effect of Human Recombinant Alkaline Phosphatase on 7-Day Creatinine Clearance in Patients With Sepsis-Associated Acute Kidney Injury: A Randomized Clinical Trial. JAMA 2018; 320:1998-2009. [PMID: 30357272 PMCID: PMC6248164 DOI: 10.1001/jama.2018.14283] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
IMPORTANCE Sepsis-associated acute kidney injury (AKI) adversely affects long-term kidney outcomes and survival. Administration of the detoxifying enzyme alkaline phosphatase may improve kidney function and survival. OBJECTIVE To determine the optimal therapeutic dose, effect on kidney function, and adverse effects of a human recombinant alkaline phosphatase in patients who are critically ill with sepsis-associated AKI. DESIGN, SETTING, AND PARTICIPANTS The STOP-AKI trial was an international (53 recruiting sites), randomized, double-blind, placebo-controlled, dose-finding, adaptive phase 2a/2b study in 301 adult patients admitted to the intensive care unit with a diagnosis of sepsis and AKI. Patients were enrolled between December 2014 and May 2017, and follow-up was conducted for 90 days. The final date of follow-up was August 14, 2017. INTERVENTIONS In the intention-to-treat analysis, in part 1 of the trial, patients were randomized to receive recombinant alkaline phosphatase in a dosage of 0.4 mg/kg (n = 31), 0.8 mg/kg (n = 32), or 1.6 mg/kg (n = 29) or placebo (n = 30), once daily for 3 days, to establish the optimal dose. The optimal dose was identified as 1.6 mg/kg based on modeling approaches and adverse events. In part 2, 1.6 mg/kg (n = 82) was compared with placebo (n = 86). MAIN OUTCOMES AND MEASURES The primary end point was the time-corrected area under the curve of the endogenous creatinine clearance for days 1 through 7, divided by 7 to provide a mean daily creatinine clearance (AUC1-7 ECC). Incidence of fatal and nonfatal (serious) adverse events ([S]AEs) was also determined. RESULTS Overall, 301 patients were enrolled (men, 70.7%; median age, 67 years [interquartile range {IQR}, 59-73]). From day 1 to day 7, median ECC increased from 26.0 mL/min (IQR, 8.8 to 59.5) to 65.4 mL/min (IQR, 26.7 to 115.4) in the recombinant alkaline phosphatase 1.6-mg/kg group vs from 35.9 mL/min (IQR, 12.2 to 82.9) to 61.9 mL/min (IQR, 22.7 to 115.2) in the placebo group (absolute difference, 9.5 mL/min [95% CI, -23.9 to 25.5]; P = .47). Fatal adverse events occurred in 26.3% of patients in the 0.4-mg/kg recombinant alkaline phosphatase group; 17.1% in the 0.8-mg/kg group, 17.4% in the 1.6-mg/kg group, and 29.5% in the placebo group. Rates of nonfatal SAEs were 21.0% for the 0.4-mg/kg recombinant alkaline phosphatase group, 14.3% for the 0.8-mg/kg group, 25.7% for the 1.6-mg/kg group, and 20.5% for the placebo group. CONCLUSIONS AND RELEVANCE Among patients who were critically ill with sepsis-associated acute kidney injury, human recombinant alkaline phosphatase compared with placebo did not significantly improve short-term kidney function. Further research is necessary to assess other clinical outcomes. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02182440.
Collapse
Affiliation(s)
- Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands
| | - Ravindra L. Mehta
- Department of Medicine, University of California, San Diego, Medical Center
| | | | - Michael Joannidis
- Department of Internal Medicine, Division of Intensive Care and Emergency Medicine, Medical University Innsbruck, Innsbruck, Austria
| | | | - John A. Kellum
- Department of Critical Care Medicine, Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mirjam Bachler
- Department of General and Surgical Critical Care Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Eric A. J. Hoste
- Intensive Care, Ghent University, Ghent, Belgium
- Clinical Research Foundation Flanders, Brussels, Belgium
| | - Oscar Hoiting
- Intensive Care, Canisius Wilhelmina Hospital, Nijmegen, the Netherlands
| | - Kenneth Krell
- Internal Medicine, Eastern Idaho Regional Medical Center, Idaho Falls
| | - Marlies Ostermann
- Guy’s and St Thomas’ Hospital, King’s College London, London, United Kingdom
| | - Wim Rozendaal
- Intensive Care, Jeroen Bosch Hospital, ‘s-Hertogenbosch, the Netherlands
| | - Miia Valkonen
- Division of Anesthesia and Intensive Care Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - David Brealey
- Division of Critical Care, University College London Hospitals National Institute for Health Research Biomedical Research Centre, London, United Kingdom
- Bloomsbury Institute of Intensive Care Medicine, University College Hospital, London, United Kingdom
| | | | - Ferhat Meziani
- Faculté de Médecine, Service de Réanimation, Université de Strasbourg, Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, Strasbourg, France
| | - Raghavan Murugan
- Department of Critical Care Medicine, Center for Critical Care Nephrology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Hilde de Geus
- Department of Intensive Care, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Didier Payen
- Unité Mixte de Recherche INSERM 1160, University Paris 7 Denis Diderot, Paris, France
- Department of Anaesthesiology and Critical Care, Hôpital Lariboisière, Assistance Publique—Hôpitaux de Paris, Paris, France
| | | | - Jacques Arend
- Clinical Department, AM-Pharma BV, Bunnik, the Netherlands
| |
Collapse
|
23
|
Bover J, Ureña P, Aguilar A, Mazzaferro S, Benito S, López-Báez V, Ramos A, daSilva I, Cozzolino M. Alkaline Phosphatases in the Complex Chronic Kidney Disease-Mineral and Bone Disorders. Calcif Tissue Int 2018; 103:111-124. [PMID: 29445837 DOI: 10.1007/s00223-018-0399-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/29/2018] [Indexed: 12/16/2022]
Abstract
Alkaline phosphatases (APs) remove the phosphate (dephosphorylation) needed in multiple metabolic processes (from many molecules such as proteins, nucleotides, or pyrophosphate). Therefore, APs are important for bone mineralization but paradoxically they can also be deleterious for other processes, such as vascular calcification and the increasingly known cross-talk between bone and vessels. A proper balance between beneficial and harmful activities is further complicated in the context of chronic kidney disease (CKD). In this narrative review, we will briefly update the complexity of the enzyme, including its different isoforms such as the bone-specific alkaline phosphatase or the most recently discovered B1x. We will also analyze the correlations and potential discrepancies with parathyroid hormone and bone turnover and, most importantly, the valuable recent associations of AP's with cardiovascular disease and/or vascular calcification, and survival. Finally, a basic knowledge of the synthetic and degradation pathways of APs promises to open new therapeutic strategies for the treatment of the CKD-Mineral and Bone Disorder (CKD-MBD) in the near future, as well as for other processes such as sepsis, acute kidney injury, inflammation, endothelial dysfunction, metabolic syndrome or, in diabetes, cardiovascular complications. However, no studies have been done using APs as a primary therapeutic target for clinical outcomes, and therefore, AP's levels cannot yet be used alone as an isolated primary target in the treatment of CKD-MBD. Nonetheless, its diagnostic and prognostic potential should be underlined.
Collapse
Affiliation(s)
- Jordi Bover
- Department of Nephrology, Fundació Puigvert, IIB Sant Pau, RedinRen, C. Cartagena, Catalonia, 340-350, Barcelona, Spain.
| | - Pablo Ureña
- Department of Nephrology and Dialysis, Clinique du Landy and Department of Renal Physiology, Necker Hospital, University of Paris Descartes, Paris, France
| | - Armando Aguilar
- Department of Nephrology, Fundació Puigvert, IIB Sant Pau, RedinRen, C. Cartagena, Catalonia, 340-350, Barcelona, Spain
| | - Sandro Mazzaferro
- Department of Cardiovascular, Respiratory, Nephrologic and Geriatric Sciences, Sapienza University of Rome, Rome, Italy
| | - Silvia Benito
- Department of Nephrology, Fundació Puigvert, IIB Sant Pau, RedinRen, C. Cartagena, Catalonia, 340-350, Barcelona, Spain
| | - Víctor López-Báez
- Department of Nephrology, Fundació Puigvert, IIB Sant Pau, RedinRen, C. Cartagena, Catalonia, 340-350, Barcelona, Spain
| | - Alejandra Ramos
- Department of Nephrology, Fundació Puigvert, IIB Sant Pau, RedinRen, C. Cartagena, Catalonia, 340-350, Barcelona, Spain
| | - Iara daSilva
- Department of Nephrology, Fundació Puigvert, IIB Sant Pau, RedinRen, C. Cartagena, Catalonia, 340-350, Barcelona, Spain
| | - Mario Cozzolino
- Laboratory of Experimental Nephrology, Renal Division,San Paolo Hospital, DiSS University of Milan, Milan, Italy
| |
Collapse
|
24
|
Lehto M, Groop PH. The Gut-Kidney Axis: Putative Interconnections Between Gastrointestinal and Renal Disorders. Front Endocrinol (Lausanne) 2018; 9:553. [PMID: 30283404 PMCID: PMC6157406 DOI: 10.3389/fendo.2018.00553] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 08/30/2018] [Indexed: 12/15/2022] Open
Abstract
Diabetic kidney disease (DKD) is a devastating condition associated with increased morbidity and premature mortality. The etiology of DKD is still largely unknown. However, the risk of DKD development and progression is most likely modulated by a combination of genetic and environmental factors. Patients with autoimmune diseases, like type 1 diabetes, inflammatory bowel disease, and celiac disease, share some genetic background. Furthermore, gastrointestinal disorders are associated with an increased risk of kidney disease, although the true mechanisms have still to be elucidated. Therefore, the principal aim of this review is to evaluate the impact of disturbances in the gastrointestinal tract on the development of renal disorders.
Collapse
Affiliation(s)
- Markku Lehto
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Abdominal Center of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
- *Correspondence: Markku Lehto
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Abdominal Center of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
25
|
Shang J, Zhao Z. Emerging role of HuR in inflammatory response in kidney diseases. Acta Biochim Biophys Sin (Shanghai) 2017; 49:753-763. [PMID: 28910975 DOI: 10.1093/abbs/gmx071] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 06/21/2017] [Indexed: 12/14/2022] Open
Abstract
Human antigen R (HuR) is a member of the embryonic lethal abnormal vision (ELAV) family which can bind to the A/U rich elements in 3' un-translated region of mRNA and regulate mRNA splicing, transportation, and stability. Unlike other members of the ELAV family, HuR is ubiquitously expressed. Early studies mainly focused on HuR function in malignant diseases. As researches proceed, more and more proofs demonstrate its relationship with inflammation. Since most kidney diseases involve pathological changes of inflammation, HuR is now suggested to play a pivotal role in glomerular nephropathy, tubular ischemia-reperfusion damage, renal fibrosis and even renal tumors. By regulating the mRNAs of target genes, HuR is causally linked to the onset and progression of kidney diseases. Reports on this topic are steadily increasing, however, the detailed function and mechanism of action of HuR are still not well understood. The aim of this review article is to summarize the present understanding of the role of HuR in inflammation in kidney diseases, and we anticipate that future research will ultimately elucidate the therapeutic value of this novel target.
Collapse
Affiliation(s)
- Jin Shang
- Nephrology Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhanzheng Zhao
- Nephrology Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
26
|
Peters E, Schirris T, van Asbeck AH, Gerretsen J, Eymael J, Ashikov A, Adjobo-Hermans MJW, Russel F, Pickkers P, Masereeuw R. Effects of a human recombinant alkaline phosphatase during impaired mitochondrial function in human renal proximal tubule epithelial cells. Eur J Pharmacol 2016; 796:149-157. [PMID: 28012971 DOI: 10.1016/j.ejphar.2016.12.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/19/2016] [Accepted: 12/21/2016] [Indexed: 01/07/2023]
Abstract
Sepsis-associated acute kidney injury is a multifactorial syndrome in which inflammation and renal microcirculatory dysfunction play a profound role. Subsequently, renal tubule mitochondria reprioritize cellular functions to prevent further damage. Here, we investigated the putative protective effects of human recombinant alkaline phosphatase (recAP) during inhibition of mitochondrial respiration in conditionally immortalized human proximal tubule epithelial cells (ciPTEC). Full inhibition of mitochondrial oxygen consumption was obtained after 24h antimycin A treatment, which did not affect cell viability. While recAP did not affect the antimycin A-induced decreased oxygen consumption and increased hypoxia-inducible factor-1α or adrenomedullin gene expression levels, the antimycin A-induced increase of pro-inflammatory cytokines IL-6 and IL-8 was attenuated. Antimycin A tended to induce the release of detrimental purines ATP and ADP, which reached statistical significance when antimycin A was co-incubated with lipopolysaccharide, and were completely converted into cytoprotective adenosine by recAP. As the adenosine A2A receptor was up-regulated after antimycin A exposure, an adenosine A2A receptor knockout ciPTEC cell line was generated in which recAP still provided protection. Together, recAP did not affect oxygen consumption but attenuated the inflammatory response during impaired mitochondrial function, an effect suggested to be mediated by dephosphorylating ATP and ADP into adenosine.
Collapse
Affiliation(s)
- Esther Peters
- Department of Intensive Care Medicine, Radboud University Medical Center, PO Box 9101, Internal Mailbox 710, 6500 HB Nijmegen, The Netherlands; Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, Internal Mailbox 710, 6500 HB Nijmegen, The Netherlands.
| | - Tom Schirris
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, Internal Mailbox 710, 6500 HB Nijmegen, The Netherlands; Radboud Institute for Mitochondrial Medicine, Radboud University Medical Center, PO Box 9101, Internal Mailbox 710, 6500 HB Nijmegen, The Netherlands.
| | - Alexander H van Asbeck
- Department of Biochemistry, Radboud University Medical Center, PO Box 9101, Internal Mailbox 710, 6500 HB Nijmegen, The Netherlands.
| | - Jelle Gerretsen
- Department of Intensive Care Medicine, Radboud University Medical Center, PO Box 9101, Internal Mailbox 710, 6500 HB Nijmegen, The Netherlands.
| | - Jennifer Eymael
- Department of Intensive Care Medicine, Radboud University Medical Center, PO Box 9101, Internal Mailbox 710, 6500 HB Nijmegen, The Netherlands.
| | - Angel Ashikov
- Department of Neurology, Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, PO Box 9101, Internal Mailbox 710, 6500 HB Nijmegen, The Netherlands.
| | - Merel J W Adjobo-Hermans
- Department of Biochemistry, Radboud University Medical Center, PO Box 9101, Internal Mailbox 710, 6500 HB Nijmegen, The Netherlands.
| | - Frans Russel
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, Internal Mailbox 710, 6500 HB Nijmegen, The Netherlands; Radboud Institute for Mitochondrial Medicine, Radboud University Medical Center, PO Box 9101, Internal Mailbox 710, 6500 HB Nijmegen, The Netherlands.
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, PO Box 9101, Internal Mailbox 710, 6500 HB Nijmegen, The Netherlands.
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, PO BOX 80082, 3508 TB Utrecht, The Netherlands.
| |
Collapse
|