1
|
Nakade Y, Iwata Y, Harada K, Sato Y, Mita M, Hamase K, Konno R, Hayashi M, Kobayashi T, Yamamura Y, Toyama T, Tajima A, Wada T. Effect of D-amino acid metabolic enzyme deficiency on cancer development-diffuse large B-cell lymphoma onset and gene expression analyses in DASPO-knockout mice. Amino Acids 2024; 57:4. [PMID: 39718666 DOI: 10.1007/s00726-024-03426-1] [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: 09/19/2024] [Accepted: 11/06/2024] [Indexed: 12/25/2024]
Abstract
The relationship between D-AA metabolic enzymes and cancer development remains unclear. We aimed to investigate this relationship using mice deficient in D-AA-related metabolic enzymes. We examined mice lacking these enzymes for approximately 900 days and the effects of altered D-AA metabolism on cancer development based on lifespan, pathological findings, and gene expression. The lifespan of female DASPO -knockout (DASPO-/-) mice was shorter than that of the other group mice; furthermore, these mice showed tumor-like masses in the liver, spleen, and small intestine. A pathological diagnosis of diffuse large B-cell lymphoma (DLBCL) was made. RNA sequencing of the liver samples showed specific alterations in the expression of 71 genes in DASPO-/- mice compared with that in wild-type B6 mice; RGS 1, MTSS1, and SMARCD 1 were identified as DLBCL-related genes. Patients with DLBCL exhibiting low DASPO expression demonstrated a shorter survival period than those showing high expression. However, the role of DASPO in DLBCL development is unclear. Therefore, future research should focus on B cells. DASPO may serve as novel biomarkers and therapeutic targets in cancer.
Collapse
Affiliation(s)
- Yusuke Nakade
- Department of Nephrology and Rheumatology, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
- Department of Clinical Laboratory, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Yasunori Iwata
- Department of Nephrology and Rheumatology, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
- Department of Clinical Laboratory, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Kenichi Harada
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Yasuharu Sato
- Department of Molecular Hematopathology, Okayama University Graduate School of Health Sciences, 2-5-1 Shikata-Chou, Kita-Ku, Okayama, 700-8558, Japan
| | - Masashi Mita
- KAGAMI Co., Ltd., 7-18 Saitobaiohiruzu Centre 308, Ibaragi-Shi, Osaka, 567-0085, Japan
| | - Kenji Hamase
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Ryuichi Konno
- Department of Pharmaceutical Sciences, International University of Health and Welfare, 2600-1 Kitakanemaru, Ohtawara, 324-8501, Japan
| | - Mayo Hayashi
- Department of Nephrology and Rheumatology, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
- Department of Clinical Laboratory, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Taku Kobayashi
- Department of Nephrology and Rheumatology, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Yuta Yamamura
- Department of Nephrology and Rheumatology, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Tadashi Toyama
- Department of Nephrology and Rheumatology, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Atsushi Tajima
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Takashi Wada
- Department of Nephrology and Rheumatology, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan.
| |
Collapse
|
2
|
Kimura T, Sakai S, Horio M, Takahara S, Ishigo S, Nakane M, Negishi E, Imoto H, Mita M, Hamase K, Higa-Maekawa Y, Kakuta Y, Mizui M, Isaka Y. Kinetic analysis of D-Alanine upon oral intake in humans. Amino Acids 2024; 56:61. [PMID: 39400632 PMCID: PMC11473621 DOI: 10.1007/s00726-024-03421-6] [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: 05/22/2024] [Accepted: 09/27/2024] [Indexed: 10/15/2024]
Abstract
D-Alanine, a rare enantiomer of alanine, can potentially alleviate the worsening of viral infections and maintain circadian rhythm. This study aimed to analyze the kinetics of D-Alanine upon oral intake. Five healthy volunteers were administered D-Alanine as a single oral dose at 11,236 or 33,708 µmoL (1-3 g). Upon intake of the lower dose, the plasma level of D-Alanine reached its peak concentration of 588.4 ± 40.9 µM with a peak time of 0.60 ± 0.06 h. The compartment model estimated the clearance of D-Alanine at 12.5 ± 0.3 L/h, or 208 ± 5 mL/min, distribution volume of 8.3 ± 0.7 L and half-life of 0.46 ± 0.04 h, suggesting a rapid clearance of D-Alanine. The peak concentration and area under the curve increased proportionally upon intake of the higher dose, while the clearance, distribution volume and half-life did not. The urinary ratio of D-Alanine per sum of D- and L-Alanine reached its peak of nearly 100%, followed by a slow decline. The peak time of the urinary ratio was 1.15 ± 0.15 h, showing a time lag of blood to urine excretion. Fractional excretion, a ratio of the clearance of a substance per a standard molecule in kidney, of D-Alanine increased from 14.0 ± 5.8% to 64.5 ± 10.3%; the latter corresponded to the urinary clearance of D-Alanine as about 77 mL/min for an adult, with a peak time of 1.90 ± 0.56 h. D-Alanine was quickly absorbed and appeared in blood, followed by urinary excretion. This kinetic analysis increases our fundamental knowledge of the oral intake of D-Alanine for the chronic dosing. Trial number: #UMIN000050865. Date of registration: 2023/6/30.
Collapse
Affiliation(s)
- Tomonori Kimura
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan.
- Department of Nephrology, Kansai Medical Hospital, Osaka, Japan.
- Kansai Medical Clinic for Renal Transplantation, Osaka, Japan.
| | - Shinsuke Sakai
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masaru Horio
- Department of Nephrology, Kansai Medical Hospital, Osaka, Japan
| | - Shiro Takahara
- Kansai Medical Clinic for Renal Transplantation, Osaka, Japan
| | | | | | | | | | | | - Kenji Hamase
- Department of Drug Discovery and Evolution, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoko Higa-Maekawa
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoichi Kakuta
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masayuki Mizui
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
3
|
Oshima M, Toyama T, Toyama T, Nakade Y, Tokumaru T, Sako K, Kajikawa S, Hayashi D, Sanada H, Yuasa T, Koshino A, Horikoshi K, Minami T, Tsuge S, Tamai A, Nakagawa S, Nishioka R, Zoshima T, Ito K, Kitajima S, Mizushima I, Hara A, Sakai N, Shimizu M, Mita M, Iwata Y, Wada T. Effects of d-alanine Intake on Amino Acid Metabolism and Kidney Function in Healthy Adults: A Multicenter, Randomized Pilot Study. Curr Dev Nutr 2024; 8:103787. [PMID: 39045146 PMCID: PMC11262164 DOI: 10.1016/j.cdnut.2024.103787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 05/17/2024] [Indexed: 07/25/2024] Open
Abstract
Background d-alanine administration prevented kidney damage in a murine acute kidney injury model. Further data are needed on the influence of d-alanine on kidney function in humans. Objective This study investigated the effects of d-alanine intake on amino acid metabolism and kidney function in healthy volunteers. Methods This multicenter pilot study randomly assigned individuals from the general Japanese population to receive 3 g or 6 g of d-alanine intake per day for 7 d in a 1:1 ratio. The primary endpoint was the mean change in plasma and urine d-alanine levels from baseline to 7 d after intake. The secondary endpoints were mean changes in kidney function and other clinical factors. Safety was assessed by evaluating adverse events and clinical parameters. Results We randomly assigned 24 participants to the 3-g (n = 12) and 6-g d-alanine (n = 12) groups. The mean baseline estimated glomerular filtration rate (eGFR) was 73 mL/min/1.73 m2. The mean plasma d-alanine concentration increased from baseline by 77.5 ± 34.3 and 192.1 ± 80.9 nmol/mL in the 3-g and 6-g d-alanine groups (both p < 0.0001), respectively, in a dose-dependent manner (between-group difference: 114.6 nmol/mL; 95% CI: 62.1-167.2; P = 0.0002). A similar increase was observed for the urine d-alanine to creatinine ratio. The mean eGFR was elevated by 5.7 ± 8.8 mL/min/1.73 m2 in the 6-g d-alanine group (P = 0.045) but did not significantly change in the 3-g d-alanine group. Nonserious adverse events were reported in 11 participants. Conclusions d-alanine intake increased plasma and urine d-alanine levels and was well tolerated in participants with normal kidney function. These results will be useful in future trials investigating the effects of d-alanine intake on kidney disease progression in patients with chronic kidney disease.This trial was registered at the UMIN Clinical Trials Registry as UMIN000051466.
Collapse
Affiliation(s)
- Megumi Oshima
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Tadashi Toyama
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
- Kouryo Clinic, Takaoka, Japan
| | | | - Yusuke Nakade
- Department of Clinical Laboratory, Kanazawa University, Kanazawa, Japan
| | - Toshiaki Tokumaru
- Department of Nutrition, Kanazawa University Hospital, Kanazawa, Japan
| | - Keisuke Sako
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Sho Kajikawa
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Daiki Hayashi
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Hajime Sanada
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Takahiro Yuasa
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Akihiko Koshino
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Keisuke Horikoshi
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Taichiro Minami
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Shunsuke Tsuge
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Akira Tamai
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Shiori Nakagawa
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Ryo Nishioka
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Takeshi Zoshima
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Kiyoaki Ito
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Shinji Kitajima
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Ichiro Mizushima
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Akinori Hara
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Norihiko Sakai
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Miho Shimizu
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | | | - Yasunori Iwata
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Takashi Wada
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
4
|
Bantounas I, Rooney KM, Lopes FM, Tengku F, Woods S, Zeef LAH, Lin IH, Kuba SY, Bates N, Hummelgaard S, Hillman KA, Cereghini S, Woolf AS, Kimber SJ. Human pluripotent stem cell-derived kidney organoids reveal tubular epithelial pathobiology of heterozygous HNF1B-associated dysplastic kidney malformations. Stem Cell Reports 2024; 19:859-876. [PMID: 38788724 PMCID: PMC11297557 DOI: 10.1016/j.stemcr.2024.04.011] [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: 02/14/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Hepatocyte nuclear factor 1B (HNF1B) encodes a transcription factor expressed in developing human kidney epithelia. Heterozygous HNF1B mutations are the commonest monogenic cause of dysplastic kidney malformations (DKMs). To understand their pathobiology, we generated heterozygous HNF1B mutant kidney organoids from CRISPR-Cas9 gene-edited human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) reprogrammed from a family with HNF1B-associated DKMs. Mutant organoids contained enlarged malformed tubules displaying deregulated cell turnover. Numerous genes implicated in Mendelian kidney tubulopathies were downregulated, and mutant tubules resisted the cyclic AMP (cAMP)-mediated dilatation seen in controls. Bulk and single-cell RNA sequencing (scRNA-seq) analyses indicated abnormal Wingless/Integrated (WNT), calcium, and glutamatergic pathways, the latter hitherto unstudied in developing kidneys. Glutamate ionotropic receptor kainate type subunit 3 (GRIK3) was upregulated in malformed mutant nephron tubules and prominent in HNF1B mutant fetal human dysplastic kidney epithelia. These results reveal morphological, molecular, and physiological roles for HNF1B in human kidney tubule differentiation and morphogenesis illuminating the developmental origin of mutant-HNF1B-causing kidney disease.
Collapse
Affiliation(s)
- Ioannis Bantounas
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK
| | - Kirsty M Rooney
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK
| | - Filipa M Lopes
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK
| | - Faris Tengku
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK
| | - Steven Woods
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK
| | - Leo A H Zeef
- Bioinformatics Core Facility, University of Manchester, Manchester, UK
| | - I-Hsuan Lin
- Bioinformatics Core Facility, University of Manchester, Manchester, UK
| | - Shweta Y Kuba
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK
| | - Nicola Bates
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK
| | - Sandra Hummelgaard
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK; Department of Biomedicine, Aarhus University, Denmark
| | - Katherine A Hillman
- Manchester Institute of Nephrology and Transplantation, Manchester University NHS Foundation Trust, Manchester, UK
| | - Silvia Cereghini
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratorial de Biologie du Développement, IBPS, UMR7622, F-75005 Paris, France
| | - Adrian S Woolf
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK; Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, UK.
| | - Susan J Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, and the Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
5
|
Hashimoto H, Takagi T, Asaeda K, Yasuda T, Kajiwara M, Sugaya T, Mizushima K, Inoue K, Uchiyama K, Kamada K, Higashimura Y, Inoue R, Naito Y, Itoh Y. D-alanine Inhibits Murine Intestinal Inflammation by Suppressing IL-12 and IL-23 Production in Macrophages. J Crohns Colitis 2024; 18:908-919. [PMID: 38165390 DOI: 10.1093/ecco-jcc/jjad217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 11/21/2023] [Accepted: 12/30/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND AND AIMS Free D-amino acids, which have different functions from L-amino acids, have recently been discovered in various tissues. However, studies on the potential interactions between intestinal inflammation and D-amino acids are limited. We examined the inhibitory effects of D-alanine on the pathogenesis of intestinal inflammation. METHODS We investigated serum D-amino acid levels in 40 patients with ulcerative colitis and 34 healthy volunteers. For 7 days [d], acute colitis was induced using dextran sulphate sodium in C57BL/6J mice. Plasma D-amino acid levels were quantified in mice with dextran sulphate sodium-induced colitis, and these animals were administered D-alanine via intraperitoneal injection. IFN-γ, IL-12p35, IL-17A, and IL-23p19 mRNA expression in the colonic mucosa was measured using real-time polymerase chain reaction [PCR]. In vitro proliferation assays were performed to assess naïve CD4+ T cell activation under Th-skewing conditions. Bone marrow cells were stimulated with mouse macrophage-colony stimulating factor to generate mouse bone marrow-derived macrophages. RESULTS Serum D-alanine levels were significantly lower in patients with ulcerative colitis than in healthy volunteers. Dextran sulphate sodium-treated mice had significantly lower plasma D-alanine levels than control mice. D-alanine-treated mice had significantly lower disease activity index than control mice. IFN-γ, IL-12p35, IL-17A, and IL-23p19 mRNA expression levels were significantly lower in D-alanine-administered mice than in control mice. D-alanine suppressed naïve T cell differentiation into Th1 cells in vitro, and inhibited the production of IL-12p35 and IL-23p19 in bone marrow-derived macrophages. CONCLUSIONS Our results suggest that D-alanine prevents dextran sulphate sodium-induced colitis in mice and suppresses IL-12p35 and IL-23p19 production in macrophages.
Collapse
Affiliation(s)
- Hikaru Hashimoto
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Tomohisa Takagi
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
- Department for Medical Innovation and Translational Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kohei Asaeda
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Takeshi Yasuda
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Mariko Kajiwara
- Department of Gastroenterology, Fukuchiyama City Hospital, Fukuchiyama, Japan
| | - Takeshi Sugaya
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Katsura Mizushima
- Department of Human Immunology and Nutrition Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ken Inoue
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Kazuhiko Uchiyama
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Kazuhiro Kamada
- Department of Gastroenterology, Matsushita Memorial Hospital, Moriguchi, Japan
| | - Yasuki Higashimura
- Department of Food Science, Ishikawa Prefectural University, Nonoichi, Japan
| | - Ryo Inoue
- Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University, Hirakata, Japan
| | - Yuji Naito
- Department of Human Immunology and Nutrition Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshito Itoh
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| |
Collapse
|
6
|
Tokumaru T, Toyama T, Nakade Y, Ogura H, Oshima M, Nakagawa S, Furuichi M, Kitajima S, Sakai N, Shimizu M, Iwata Y, Wada T. Design and rationale for an open-label, randomized, controlled pilot trial to evaluate the changes in blood uremic toxins in patients with chronic kidney disease by dietary therapy with sake lees. Clin Exp Nephrol 2024; 28:440-446. [PMID: 38340247 PMCID: PMC11033224 DOI: 10.1007/s10157-023-02450-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/18/2023] [Indexed: 02/12/2024]
Abstract
BACKGROUND Patients with chronic kidney disease (CKD) reportedly show dysbiosis, which is the imbalance of gut microbiome. Dysbiosis increases the uremic toxin level in the intestine, and uremic toxins transfer into the blood, causing CKD progression. Sake lees, a traditional Japanese fermented food, may help reduce uremic toxins by altering the gut microbiome. Additionally, D-alanine, which is present in sake lees, may have a renoprotective effect. The present pilot study aims to evaluate the effect of adding sake lees to the standard CKD dietary therapy in reducing blood uremic toxins. METHODS This pilot study is a single-center, open-label, randomized controlled trial. Twenty-four patients with CKD will be enrolled and allocated 1:1 to the intervention and control groups. The intervention group will receive standard CKD dietary therapy with an additional intake of 50 g of sake lees per day for 8 weeks, whereas the control group will only receive standard CKD dietary therapy. The primary endpoint is the change in serum indoxyl sulfate after 8 weeks. The secondary endpoint is the plasma D-alanine and fecal microbiome changes. CONCLUSION This pilot study provides insight into the development of a new diet focused on gut microbiome and D-amino acids in patients with CKD. CLINICAL TRIAL REGISTRATION This protocol was approved by the Clinical Trial Review Board of Kanazawa University Hospital on October 27, 2022 (2022-001 [6139]) and available to the public on the website of the Japan Registry of Clinical Trials on November 22, 2022 (jRCT1040220095).
Collapse
Affiliation(s)
- Toshiaki Tokumaru
- Department of Nephrology and Rheumatology, Kanazawa University, Takara-Machi 13-1, Kanazawa City, Ishikawa, 920-8641, Japan
- Department of Nutrition Management, Kanazawa University Hospital, Kanazawa, Japan
| | - Tadashi Toyama
- Department of Nephrology and Rheumatology, Kanazawa University, Takara-Machi 13-1, Kanazawa City, Ishikawa, 920-8641, Japan.
- Innovative Clinical Research Center, Kanazawa University, Kanazawa, Japan.
| | - Yusuke Nakade
- Department of Nephrology and Rheumatology, Kanazawa University, Takara-Machi 13-1, Kanazawa City, Ishikawa, 920-8641, Japan
| | - Hisayuki Ogura
- Department of Nephrology and Rheumatology, Kanazawa University, Takara-Machi 13-1, Kanazawa City, Ishikawa, 920-8641, Japan
| | - Megumi Oshima
- Department of Nephrology and Rheumatology, Kanazawa University, Takara-Machi 13-1, Kanazawa City, Ishikawa, 920-8641, Japan
| | - Shiori Nakagawa
- Department of Nephrology and Rheumatology, Kanazawa University, Takara-Machi 13-1, Kanazawa City, Ishikawa, 920-8641, Japan
| | - Motoe Furuichi
- Department of Nutrition Management, Kanazawa University Hospital, Kanazawa, Japan
| | - Shinji Kitajima
- Department of Nephrology and Rheumatology, Kanazawa University, Takara-Machi 13-1, Kanazawa City, Ishikawa, 920-8641, Japan
| | - Norihiko Sakai
- Department of Nephrology and Rheumatology, Kanazawa University, Takara-Machi 13-1, Kanazawa City, Ishikawa, 920-8641, Japan
| | - Miho Shimizu
- Department of Nephrology and Rheumatology, Kanazawa University, Takara-Machi 13-1, Kanazawa City, Ishikawa, 920-8641, Japan
| | - Yasunori Iwata
- Department of Nephrology and Rheumatology, Kanazawa University, Takara-Machi 13-1, Kanazawa City, Ishikawa, 920-8641, Japan
- Division of Infection Control, Kanazawa University Hospital, Kanazawa, Japan
| | - Takashi Wada
- Department of Nephrology and Rheumatology, Kanazawa University, Takara-Machi 13-1, Kanazawa City, Ishikawa, 920-8641, Japan
| |
Collapse
|
7
|
Yang K, Liu J, He T, Dong W. Caffeine and neonatal acute kidney injury. Pediatr Nephrol 2024; 39:1355-1367. [PMID: 37665410 DOI: 10.1007/s00467-023-06122-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023]
Abstract
Acute kidney injury is one of the most threatening diseases in neonates, with complex pathogenesis and limited treatment options. Caffeine is a commonly used central nervous system stimulant for treating apnea in preterm infants. There is compelling evidence that caffeine may have potential benefits for preventing neonatal acute kidney injury, but comprehensive reports are lacking in this area. Hence, this review aims to provide a summary of clinical data on the potential benefits of caffeine in improving neonatal acute kidney injury. Additionally, it delves into the molecular mechanisms underlying caffeine's effects on acute kidney injury, with a focus on various aspects such as oxidative stress, adenosine receptors, mitochondrial dysfunction, endoplasmic reticulum stress, inflammasome, autophagy, p53, and gut microbiota. The ultimate goal of this review is to provide information for healthcare professionals regarding the link between caffeine and neonatal acute kidney injury and to identify gaps in our current understanding.
Collapse
Affiliation(s)
- Kun Yang
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China
| | - Jinjing Liu
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China
| | - Ting He
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China
| | - Wenbin Dong
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China.
| |
Collapse
|
8
|
Wiriyasermkul P, Moriyama S, Suzuki M, Kongpracha P, Nakamae N, Takeshita S, Tanaka Y, Matsuda A, Miyasaka M, Hamase K, Kimura T, Mita M, Sasabe J, Nagamori S. <sc>A</sc> multi-hierarchical approach reveals <sc>d</sc>-serine as a hidden substrate of sodium-coupled monocarboxylate transporters. eLife 2024; 12:RP92615. [PMID: 38650461 PMCID: PMC11037918 DOI: 10.7554/elife.92615] [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] [Indexed: 04/25/2024] Open
Abstract
Transporter research primarily relies on the canonical substrates of well-established transporters. This approach has limitations when studying transporters for the low-abundant micromolecules, such as micronutrients, and may not reveal physiological functions of the transporters. While d-serine, a trace enantiomer of serine in the circulation, was discovered as an emerging biomarker of kidney function, its transport mechanisms in the periphery remain unknown. Here, using a multi-hierarchical approach from body fluids to molecules, combining multi-omics, cell-free synthetic biochemistry, and ex vivo transport analyses, we have identified two types of renal d-serine transport systems. We revealed that the small amino acid transporter ASCT2 serves as a d-serine transporter previously uncharacterized in the kidney and discovered d-serine as a non-canonical substrate of the sodium-coupled monocarboxylate transporters (SMCTs). These two systems are physiologically complementary, but ASCT2 dominates the role in the pathological condition. Our findings not only shed light on renal d-serine transport, but also clarify the importance of non-canonical substrate transport. This study provides a framework for investigating multiple transport systems of various trace micromolecules under physiological conditions and in multifactorial diseases.
Collapse
Affiliation(s)
- Pattama Wiriyasermkul
- Center for SI Medical Research, The Jikei University School of MedicineTokyoJapan
- Department of Laboratory Medicine, The Jikei University School of MedicineTokyoJapan
- Department of Collaborative Research for Biomolecular Dynamics, Nara Medical UniversityNaraJapan
| | - Satomi Moriyama
- Department of Collaborative Research for Biomolecular Dynamics, Nara Medical UniversityNaraJapan
| | - Masataka Suzuki
- Department of Pharmacology, Keio University School of MedicineTokyoJapan
| | - Pornparn Kongpracha
- Center for SI Medical Research, The Jikei University School of MedicineTokyoJapan
- Department of Laboratory Medicine, The Jikei University School of MedicineTokyoJapan
| | - Nodoka Nakamae
- Department of Collaborative Research for Biomolecular Dynamics, Nara Medical UniversityNaraJapan
| | - Saki Takeshita
- Department of Collaborative Research for Biomolecular Dynamics, Nara Medical UniversityNaraJapan
| | - Yoko Tanaka
- Department of Collaborative Research for Biomolecular Dynamics, Nara Medical UniversityNaraJapan
| | - Akina Matsuda
- Department of Pharmacology, Keio University School of MedicineTokyoJapan
| | - Masaki Miyasaka
- Center for SI Medical Research, The Jikei University School of MedicineTokyoJapan
- Department of Laboratory Medicine, The Jikei University School of MedicineTokyoJapan
| | - Kenji Hamase
- Graduate School of Pharmaceutical Sciences, Kyushu UniversityFukuokaJapan
| | - Tomonori Kimura
- KAGAMI Project, National Institutes of Biomedical Innovation, Health and NutritionOsakaJapan
- Reverse Translational Research Project, Center for Rare Disease Research, National Institutes of Biomedical Innovation, Health and NutritionOsakaJapan
| | | | - Jumpei Sasabe
- Department of Pharmacology, Keio University School of MedicineTokyoJapan
| | - Shushi Nagamori
- Center for SI Medical Research, The Jikei University School of MedicineTokyoJapan
- Department of Laboratory Medicine, The Jikei University School of MedicineTokyoJapan
- Department of Collaborative Research for Biomolecular Dynamics, Nara Medical UniversityNaraJapan
| |
Collapse
|
9
|
Rakhmetova KK, Mishina ES, Bobyntsev II, Bezhin AI, Vorvul AO. Effects of Gly-His-Lys-D-Ala Peptide on Skin Wound Regeneration Processes. Bull Exp Biol Med 2024; 176:411-416. [PMID: 38345677 DOI: 10.1007/s10517-024-06035-w] [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: 05/31/2023] [Indexed: 02/22/2024]
Abstract
We evaluated the effects of the Gly-His-Lys-D-Ala peptide in a dose 0.5 μg/kg on skin wound regeneration in male Wistar rats (n=80) after initial surgical debridement when administered intracutaneously around the site of injury. Histological (severity of the inflammatory reaction, formation of granulation tissue, and epithelialization terms) and morphometric (number of fibroblastic cells, macrophages, granulocytes, and lymphocytes) studies were performed on autopsy specimens on days 3, 7, 10, and 30 of the experiment. Daily intracutaneous injection of the peptide resulted in an increase in the number of fibroblastic cells and macrophages, as well as in a decrease in the number of granulocytes against the background of active wound contraction on day 30 of the experiment. Thus, Gly-His-Lys-D-Ala alleviated the inflammatory reaction and promoted the regenerative processes.
Collapse
Affiliation(s)
- K K Rakhmetova
- Kursk State Medical University, Ministry of Health of the Russian Federation, Kursk, Russia
| | - E S Mishina
- Kursk State Medical University, Ministry of Health of the Russian Federation, Kursk, Russia
| | - I I Bobyntsev
- Kursk State Medical University, Ministry of Health of the Russian Federation, Kursk, Russia.
| | - A I Bezhin
- Kursk State Medical University, Ministry of Health of the Russian Federation, Kursk, Russia
| | - A O Vorvul
- Kursk State Medical University, Ministry of Health of the Russian Federation, Kursk, Russia
| |
Collapse
|
10
|
Yang K, Du G, Liu J, Zhao S, Dong W. Gut microbiota and neonatal acute kidney injury biomarkers. Pediatr Nephrol 2023; 38:3529-3547. [PMID: 36997773 DOI: 10.1007/s00467-023-05931-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/28/2023] [Accepted: 02/28/2023] [Indexed: 04/01/2023]
Abstract
One of the most frequent issues in newborns is acute kidney injury (AKI), which can lengthen their hospital stay or potentially raise their chance of dying. The gut-kidney axis establishes a bidirectional interplay between gut microbiota and kidney illness, particularly AKI, and demonstrates the importance of gut microbiota to host health. Since the ability to predict neonatal AKI using blood creatinine and urine output as evaluation parameters is somewhat constrained, a number of interesting biomarkers have been developed. There are few in-depth studies on the relationships between these neonatal AKI indicators and gut microbiota. In order to gain fresh insights into the gut-kidney axis of neonatal AKI, this review is based on the gut-kidney axis and describes relationships between gut microbiota and neonatal AKI biomarkers.
Collapse
Affiliation(s)
- Kun Yang
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China
| | - Guoxia Du
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China
| | - Jinjing Liu
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China
| | - Shuai Zhao
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China
| | - Wenbin Dong
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China.
| |
Collapse
|
11
|
Kimura T, Sakai S, Isaka Y. D-Serine as a sensor and effector of the kidney. Clin Exp Nephrol 2023; 27:891-900. [PMID: 37498348 PMCID: PMC10582142 DOI: 10.1007/s10157-023-02384-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/11/2023] [Indexed: 07/28/2023]
Abstract
D-Serine, a rare enantiomer of serine, is a biomarker of kidney disease and function. The level of D-serine in the human body is precisely regulated through the urinary clearance of the kidney, and its clearance serves as a new measure of glomerular filtration rate with a lower bias than creatinine clearance. D-Serine also has a direct effect on the kidneys and mediates the cellular proliferation of tubular cells via mTOR signaling and induces kidney remodeling as a compensatory reaction to the loss of kidney mass. In living kidney donors, the removal of the kidney results in an increase in blood D-serine level, which in turn accelerates kidney remodeling and augments kidney clearance, thus reducing blood levels of D-serine. This feedback system strictly controls D-serine levels in the body. The function of D-serine as a biomarker and modulator of kidney function will be the basis of precision medicine for kidney diseases.
Collapse
Affiliation(s)
- Tomonori Kimura
- Reverse Translational Research Project, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Saito-Asagi 7-6-8, Ibaraki, Osaka, 5670085, Japan.
- KAGAMI Project, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Saito-Asagi 7-6-8, Ibaraki, Osaka, 5670085, Japan.
- Department of Nephrology, Osaka University Graduate School of Medicine, Yamada-oka 2-2, Suita, Osaka, 5650871, Japan.
| | - Shinsuke Sakai
- Reverse Translational Research Project, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Saito-Asagi 7-6-8, Ibaraki, Osaka, 5670085, Japan
- KAGAMI Project, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Saito-Asagi 7-6-8, Ibaraki, Osaka, 5670085, Japan
- Department of Nephrology, Osaka University Graduate School of Medicine, Yamada-oka 2-2, Suita, Osaka, 5650871, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Yamada-oka 2-2, Suita, Osaka, 5650871, Japan.
| |
Collapse
|
12
|
Oshima M, Toyama T, Nakade Y, Yomogida D, Yuasa T, Horikoshi K, Minami T, Ogura H, Nakagawa S, Miyagawa T, Kitajima S, Hara A, Sakai N, Shimizu M, Mita M, Kinoshita M, Nakada M, Kikuchi M, Iwata Y, Wada T. Association Between Risperidone Use and Kidney Function Decline in Patients with Schizophrenia: A Retrospective Cohort Study. Clin Ther 2023; 45:889-893. [PMID: 37487866 DOI: 10.1016/j.clinthera.2023.07.002] [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: 01/30/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/26/2023]
Abstract
PURPOSE Several D-amino acids have been shown to be protective against kidney injury in mice. Risperidone, a currently used atypical antipsychotic agent for schizophrenia, is also known to inhibit the activity of D-amino acid oxidase, which degrades certain D-amino acids. Based on the hypothesis that risperidone would prevent kidney disease progression, this study investigated the association between risperidone use and kidney function decline in patients with schizophrenia. METHODS This retrospective cohort study included patients who were diagnosed with schizophrenia and had data available from two or more serum creatinine measurements between April 1, 2010, and March 31, 2020. Patients who used risperidone for at least 30 days were included in the risperidone group, whereas those who had no record of risperidone use were included in the control group. Cox regression models were used to evaluate the risk for 40% decline in estimated glomerular filtration rate (eGFR) in patients treated with risperidone compared to that in the control group. FINDINGS Overall, 212 patients used risperidone and 1468 patients had no record of risperidone use. The mean age was 55 years, 759 (45%) of the patients were male, and the mean eGFR at baseline was 88 mL/min/1.73 m2. The mean age in the risperidone group was less than that in the control group (52 vs 56 years); other baseline characteristics were comparable between the two groups. During a mean follow-up of 1.6 years, 267 patients (16%) had a 40% eGFR decline. The incidence rate of 40% eGFR decline was lower in the risperidone group than in the control group (60 vs 104 per 1000 person-years). After adjustment for baseline age, sex, and eGFR, risperidone use was associated with a decreased risk for 40% eGFR decline (hazard ratio = 0.54; 95% CI, 0.33-0.87; P = 0.01). IMPLICATIONS Risperidone use may be associated with decreased risk for kidney function decline in patients with schizophrenia. Further studies are warranted to validate these findings.
Collapse
Affiliation(s)
| | | | - Yusuke Nakade
- Department of Nephrology and Laboratory Medicine; Department of Clinical Laboratory Medicine
| | | | | | | | | | | | | | | | | | - Akinori Hara
- Department of Nephrology and Laboratory Medicine
| | | | - Miho Shimizu
- Department of Nephrology and Laboratory Medicine
| | | | | | | | - Mitsuru Kikuchi
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, and the; Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | | | - Takashi Wada
- Department of Nephrology and Laboratory Medicine
| |
Collapse
|
13
|
Chulanova AA, Smakhtin MY, Bobyntsev II, Mishina ES, Artyushkova EB, Smakhtina AM. Reparative and Antioxidant Effects of New Analogues of Immunomodulator Thymogen in Experimental Model of Liver Damage. Bull Exp Biol Med 2023; 175:700-703. [PMID: 37861903 DOI: 10.1007/s10517-023-05929-5] [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: 03/07/2023] [Indexed: 10/21/2023]
Abstract
We studied the reparative and antioxidant effects of Thymogen and its new structural analogues obtained by binding amino acid D-Ala to the N- or C-end of the peptide molecule in acute toxic hepatopathy. Intragastric administration of carbon tetrachloride for 5 days caused the development of fat degeneration of hepatocytes, a decrease in catalase activity, and an increase in malondialdehyde concentration. Administration of peptides suppressed oxidative peroxidation and stimulated reparative regeneration of hepatocytes; Thymogen analogues produced more pronounced hepatotropic and antioxidant effects than Thymogen. Inclusion of D-Ala enhanced the effect of Thymogen on the processes of regeneration in hepatocytes and the antioxidant effect under conditions of acute carbon tetrachloride hepatopathy. The highest efficiency was achieved when the amino acid was added to the C-end of the molecule.
Collapse
Affiliation(s)
- A A Chulanova
- Kursk State Medical University, Ministry of Health of the Russian Federation, Kursk, Russia
| | - M Yu Smakhtin
- Kursk State Medical University, Ministry of Health of the Russian Federation, Kursk, Russia.
| | - I I Bobyntsev
- Kursk State Medical University, Ministry of Health of the Russian Federation, Kursk, Russia
| | - E S Mishina
- Kursk State Medical University, Ministry of Health of the Russian Federation, Kursk, Russia
| | - E B Artyushkova
- Kursk State Medical University, Ministry of Health of the Russian Federation, Kursk, Russia
| | - A M Smakhtina
- Kursk State Medical University, Ministry of Health of the Russian Federation, Kursk, Russia
| |
Collapse
|
14
|
Jud P, Meinitzer A, Strohmaier H, Arefnia B, Wimmer G, Obermayer-Pietsch B, Foris V, Kovacs G, Odler B, Moazedi-Fürst F, Brodmann M, Hafner F. Association of amino acids and parameters of bone metabolism with endothelial dysfunction and vasculopathic changes in limited systemic sclerosis. Front Med (Lausanne) 2023; 10:1193121. [PMID: 37425312 PMCID: PMC10327605 DOI: 10.3389/fmed.2023.1193121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/07/2023] [Indexed: 07/11/2023] Open
Abstract
Objectives Pathways contributing to endothelial dysfunction in patients with limited cutaneous systemic sclerosis (lcSSc) are largely unknown. The aim of this study was to investigate potential associations of amino acids and parameters of bone metabolism with endothelial dysfunction and vasculopathy-related changes in patients with lcSSc and early-stage vasculopathy. Methods Amino acids, calciotropic parameters, including 25-hydroxyvitamin D and parathyroid hormone (PTH), and bone turnover parameters, including osteocalcin and N-terminal peptide of procollagen-3 (P3NP), were measured in 38 lcSSc patients and 38 controls. Endothelial dysfunction was assessed by biochemical parameters, pulse-wave analysis, flow-mediated and nitroglycerine-mediated dilation. Additionally, vasculopathy-related and SSc-specific clinical changes including capillaroscopic, skin, renal, pulmonary, gastrointestinal and periodontal parameters were recorded. Results No significant differences in amino acids, calciotropic and bone turnover parameters were observed between lcSSc patients and controls. In patients with lcSSc, several significant correlations were found between selected amino acids, parameters of endothelial dysfunction, vasculopathy-related and SSc-specific clinical changes (all with p < 0.05). In addition, significant correlations were observed between PTH and 25-hydroxyvitamin D with homoarginine, and between osteocalcin, PTH and P3NP with modified Rodnan skin score and selected periodontal parameters (all with p < 0.05). Vitamin D deficiency defined as 25-hydroxyvitamin D < 20 ng/ml was associated with the presence of puffy finger (p = 0.046) and early pattern (p = 0.040). Conclusion Selected amino acids may affect endothelial function and may be associated to vasculopathy-related and clinical changes in lcSSc patients, while the association with parameters of bone metabolism seems to be minor.
Collapse
Affiliation(s)
- Philipp Jud
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Andreas Meinitzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Heimo Strohmaier
- Center of Medical Research (ZMF), Medical University of Graz, Graz, Austria
| | - Behrouz Arefnia
- Division of Restorative Dentistry, Endodontics, Periodontology and Prosthodontics, Department of Dental Medicine and Oral Health, Medical University of Graz, Graz, Austria
| | - Gernot Wimmer
- Division of Restorative Dentistry, Endodontics, Periodontology and Prosthodontics, Department of Dental Medicine and Oral Health, Medical University of Graz, Graz, Austria
| | - Barbara Obermayer-Pietsch
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Endocrinology Lab Platform, Medical University of Graz, Graz, Austria
| | - Vasile Foris
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Gabor Kovacs
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Balazs Odler
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Florentine Moazedi-Fürst
- Division of Rheumatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Marianne Brodmann
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Franz Hafner
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
15
|
Matsuura R, Doi K, Rabb H. Acute kidney injury and distant organ dysfunction-network system analysis. Kidney Int 2023; 103:1041-1055. [PMID: 37030663 DOI: 10.1016/j.kint.2023.03.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/24/2023] [Accepted: 03/24/2023] [Indexed: 04/10/2023]
Abstract
Acute kidney injury (AKI) occurs in about half of critically ill patients and associates with high in-hospital mortality, increased long-term mortality post-discharge and subsequent progression to chronic kidney disease. Numerous clinical studies have shown that AKI is often complicated by dysfunction of distant organs, which is a cause of the high mortality associated with AKI. Experimental studies have elucidated many mechanisms of AKI-induced distant organ injury, which include inflammatory cytokines, oxidative stress and immune responses. This review will provide an update on evidence of organ crosstalk and potential therapeutics for AKI-induced organ injuries, and present the new concept of a systemic organ network to balance homeostasis and inflammation that goes beyond kidney-crosstalk with a single distant organ.
Collapse
Affiliation(s)
- Ryo Matsuura
- Department of Nephrology and Endocrinology, the University of Tokyo Hospital
| | - Kent Doi
- Department of Emergency and Critical Care Medicine, the University of Tokyo Hospital.
| | - Hamid Rabb
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine
| |
Collapse
|
16
|
Nakade Y, Iwata Y, Sakai N, Mita M, Nakane M, Hamase K, Suda W, Toyama T, Kitajima S, Hara A, Shimizu M, Ogushi C, Furuichi K, Koshino Y, Morita H, Hattori M, Wada T. Increased levels of oral Streptococcus-derived D-alanine in patients with chronic kidney disease and diabetes mellitus. Sci Rep 2022; 12:21773. [PMID: 36526888 PMCID: PMC9758232 DOI: 10.1038/s41598-022-26175-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
The number of patients on hemodialysis is increasing globally; diabetes mellitus (DM) complications is the major cause of hemodialysis in patients with chronic kidney disease (CKD). The D-amino acid (AA) profile is altered in patients with CKD; however, it has not been studied in patients with CKD and DM. Furthermore, bacteria responsible for altering the D-AA profile are not well understood. Therefore, we examined the D-AA profiles and associated bacteria in patients with CKD, with and without DM. We enrolled 12 healthy controls and 54 patients with CKD, with and without DM, and determined their salivary, stool, plasma, and urine chiral AA levels using two-dimensional high-performance liquid chromatography. We performed 16S rRNA gene sequencing analysis of the oral and gut microbiota to determine the association between the abundance of bacterial species and D-AA levels. Plasma D-alanine and D-serine levels were higher in patients with CKD than in healthy adults (p < 0.01), and plasma D-alanine levels were higher in patients with CKD and DM than in those without DM. The abundance of salivary Streptococcus, which produced D-alanine, increased in patients with CKD and DM and was positively correlated with plasma D-alanine levels. Patients with CKD and DM had unique oral microbiota and D-alanine profiles. Plasma D-alanine is a potential biomarker for patients with CKD and DM.
Collapse
Affiliation(s)
- Yusuke Nakade
- grid.9707.90000 0001 2308 3329Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan ,grid.9707.90000 0001 2308 3329Department of Clinical Laboratory, Kanazawa University, 13-1 Takara-machi, Kanazawa, Japan
| | - Yasunori Iwata
- grid.9707.90000 0001 2308 3329Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan ,grid.9707.90000 0001 2308 3329Division of Infection Control, Kanazawa University, 13-1 Takara-machi, Kanazawa, Japan
| | - Norihiko Sakai
- grid.9707.90000 0001 2308 3329Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan ,grid.9707.90000 0001 2308 3329Division of Blood Purification, Kanazawa University, 13-1 Takara-machi, Kanazawa, Japan
| | - Masashi Mita
- grid.511730.1KAGAMI Co., Ltd., 7-18 Saitobaiohiruzu Center 308, Ibaragi, Osaka Japan
| | - Maiko Nakane
- grid.511730.1KAGAMI Co., Ltd., 7-18 Saitobaiohiruzu Center 308, Ibaragi, Osaka Japan
| | - Kenji Hamase
- grid.177174.30000 0001 2242 4849Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Japan
| | - Wataru Suda
- grid.509459.40000 0004 0472 0267RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa Japan ,grid.26999.3d0000 0001 2151 536XGraduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba Japan
| | - Tadashi Toyama
- grid.9707.90000 0001 2308 3329Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| | - Shinji Kitajima
- grid.9707.90000 0001 2308 3329Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| | - Akinori Hara
- grid.9707.90000 0001 2308 3329Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| | - Miho Shimizu
- grid.9707.90000 0001 2308 3329Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| | - Chikako Ogushi
- grid.9707.90000 0001 2308 3329Department of Clinical Laboratory, Kanazawa University, 13-1 Takara-machi, Kanazawa, Japan
| | - Kengo Furuichi
- grid.411998.c0000 0001 0265 5359Department of Nephrology, Kanazawa Medical University, 1-1 Uchinada, Kahoku, Ishikawa Japan
| | - Yoshitaka Koshino
- Department of Internal Medicine, Mizuho Hospital, 422-1 Tsubata, Kahoku, Ishikawa Japan
| | - Hidetoshi Morita
- grid.261356.50000 0001 1302 4472Graduate School of Environmental and Life Science, Okayama University, 1-1-1 Tsushima-naka, Okayama, Japan
| | - Masahira Hattori
- grid.509459.40000 0004 0472 0267RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa Japan ,grid.26999.3d0000 0001 2151 536XGraduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba Japan
| | - Takashi Wada
- grid.9707.90000 0001 2308 3329Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| |
Collapse
|
17
|
Promising Application of D-Amino Acids toward Clinical Therapy. Int J Mol Sci 2022; 23:ijms231810794. [PMID: 36142706 PMCID: PMC9503604 DOI: 10.3390/ijms231810794] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/25/2022] Open
Abstract
The versatile roles of D-amino acids (D-AAs) in foods, diseases, and organisms, etc., have been widely reported. They have been regarded, not only as biomarkers of diseases but also as regulators of the physiological function of organisms. Over the past few decades, increasing data has revealed that D-AAs have great potential in treating disease. D-AAs also showed overwhelming success in disengaging biofilm, which might provide promise to inhibit microbial infection. Moreover, it can effectively restrain the growth of cancer cells. Herein, we reviewed recent reports on the potential of D-AAs as therapeutic agents for treating neurological disease or tissue/organ injury, ameliorating reproduction function, preventing biofilm infection, and inhibiting cancer cell growth. Additionally, we also reviewed the potential application of D-AAs in drug modification, such as improving biostability and efficiency, which has a better effect on therapy or diagnosis.
Collapse
|