1
|
Zhang C, Wu Z, Song Y, Jin X, Hu J, Huang C, Zhou J, Lian J. Exploring diagnostic biomarkers of type 2 cardio-renal syndrome based on secreted proteins and bioinformatics analysis. Sci Rep 2024; 14:24612. [PMID: 39427047 PMCID: PMC11490509 DOI: 10.1038/s41598-024-75580-1] [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: 06/12/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
Chronic heart failure (CHF) can induce chronic kidney disease (CKD), called type 2 cardio-renal syndrome (CRS2). The mechanism is not completely clear, and there is a lack of early warning biomarkers of CKD in the context of CHF. Two CKD, one CHF-PBMC and four CHF-cardiac tissue expression profile datasets were obtained from GEO database. Differential expression analysis and WGCNA were used to detect CKD key genes and CHF-related secreted proteins. Protein-protein interaction (PPI), functional enrichment, and cMAP analysis reveal potential mechanisms and drugs of CHF-related CKD. Five machine learning algorithms were used to screen candidate biomarkers, construct a diagnostic nomogram for CKD and validate it in two external cohorts. Clinical serum samples were collected in our hospital to evaluate the correlation and diagnostic value of biomarkers and CKD. 225 CKD key genes and 316 CHF-related secreted proteins were identified. Four key subgroups, including 204 genes, were identified as CRS2-related pathogenic genes by PPI analysis. Enrichment analysis revealed that the identified subgroups exhibited significant enrichment in cytokine action, immune responses, and inflammatory processes. The cMAP analysis highlighted metiradone as a drug with greater potential for therapeutic intervention for CRS2. Utilizing five machine learning algorithms, three hub genes (CD48, COL3A1, LOXL1) were pinpointed as potential biomarkers for CKD, and a nomogram model was constructed. Receiver operating characteristic analysis demonstrated that the nomogram's area under the curve (AUC) exceeded 0.80 in both the CKD combined dataset and two external cohorts. In addition, the three biomarkers were significantly correlated with the glomerular filtration rate, and the AUC of the model predicting disease progression was 0.944. Furthermore, analysis of immune cell infiltration indicated a correlation between the three biomarkers and the infiltration fraction of macrophages, neutrophils, and other immune cells in CKD. Our clinical cohort validated the expression patterns of three biomarkers in serum, and the diagnostic model achieved an AUC of 0.876. CHF has the potential to facilitate the progression of CKD via the release of cardiac and PBMC secreted proteins. Furthermore, CD48, COL3A1, and LOXL1 have been identified as potential biomarkers for the detection of CKD.
Collapse
Affiliation(s)
- Chuanjing Zhang
- The Affiliated Lihuili Hospital of Ningbo University, Health Science Center, Ningbo University, No.57, Xingning Road, Ningbo, 315040, Zhejiang Province, China
| | - Zhuonan Wu
- Department of Cardiology, Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Yongfei Song
- The Affiliated Lihuili Hospital of Ningbo University, Health Science Center, Ningbo University, No.57, Xingning Road, Ningbo, 315040, Zhejiang Province, China
| | - Xiaojun Jin
- The Affiliated Lihuili Hospital of Ningbo University, Health Science Center, Ningbo University, No.57, Xingning Road, Ningbo, 315040, Zhejiang Province, China
| | - Jiale Hu
- The Affiliated Lihuili Hospital of Ningbo University, Health Science Center, Ningbo University, No.57, Xingning Road, Ningbo, 315040, Zhejiang Province, China
| | - Chen Huang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Jianqing Zhou
- The Affiliated Lihuili Hospital of Ningbo University, Health Science Center, Ningbo University, No.57, Xingning Road, Ningbo, 315040, Zhejiang Province, China
| | - Jiangfang Lian
- The Affiliated Lihuili Hospital of Ningbo University, Health Science Center, Ningbo University, No.57, Xingning Road, Ningbo, 315040, Zhejiang Province, China.
| |
Collapse
|
2
|
Sugiura T, Doke T, Tanaka A, Sato Y, Maeda K, Furuhashi K, Kato N, Kosugi T, Maruyama S. MPO-ANCA-positive rapidly progressive glomerulonephritis after COVID-19 vaccination during treatment of plaque psoriasis with bimekizumab. CEN Case Rep 2024:10.1007/s13730-024-00927-6. [PMID: 39297892 DOI: 10.1007/s13730-024-00927-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 08/26/2024] [Indexed: 09/21/2024] Open
Abstract
A 75-year-old man presented with MPO-ANCA-positive rapidly progressive glomerulonephritis after COVID-19 vaccination during the treatment of plaque psoriasis vulgaris with bimekizumab. Bimekizumab, an anti-IL17 monoclonal antibody, was regularly administered to control the activity of plaque psoriasis. After receiving the sixth COVID-19 vaccine, his kidney function rapidly declined over the course of weeks. Urinalysis showed microscopic hematuria and proteinuria with deformed red blood cells and granular cast. The immunology test was positive for MPO-ANCA. The patient was clinically diagnosed with MPO-ANCA-associated glomerulonephritis. As the patient lost his appetite and developed lower extremity edema with low eGFR (< 15 ml/min/1.73m2) on admission day, hemodialysis induction was initiated along with methylprednisolone pulse, followed by oral prednisolone. The kidney function and urine volume were improved in response to immunosuppressive therapy, and withdrawal from hemodialysis was considered. However, the patient developed a catheter infection due to methicillin-sensitive Staphylococcus aureus 2 weeks after the initial prednisolone treatment, causing a decline in kidney function. Antibiotics treatment for the catheter infection was effective, but kidney function remained low, resulting in dependence on regular hemodialysis. COVID-19 vaccination provides significant improvement in overall prognosis; however, there have been reports of kidney function decline and exacerbation of hematuria in patients with IgA nephropathy following vaccination. The incidence of MPO-ANCA-associated glomerulonephritis after COVID-19 vaccination was rare. Data accumulation is warranted to understand the risk factors for secondary MPO-ANCA glomerulonephritis after COVID-19 vaccination. Regular monitoring of urinalysis and kidney function after COVID-19 vaccination is recommended in patients with psoriasis vulgaris treated with IL17 monoclonal antibodies.
Collapse
Affiliation(s)
- Takuya Sugiura
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomohito Doke
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Akihito Tanaka
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuka Sato
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kayaho Maeda
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuhiro Furuhashi
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Noritoshi Kato
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoki Kosugi
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
3
|
Ruiz-Pacheco JA, Reyes-Martínez JE, Gómez-Navarro B, Castillo-Díaz LA, Portilla de Buen E. Leptospirosis: A dual threat - predisposing risk for renal transplant and trigger for renal transplant dysfunction. Hum Immunol 2024; 85:110835. [PMID: 38972268 DOI: 10.1016/j.humimm.2024.110835] [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: 01/05/2024] [Revised: 05/29/2024] [Accepted: 06/24/2024] [Indexed: 07/09/2024]
Abstract
Leptospirosis (LTPS) is a bacterial infection that affects humans, often with mild or no symptoms. It is estimated that approximately 10 % of patients with LTPS may experience multi-organ dysfunction, including renal abnormalities. In regions where LTPS is widespread, a considerable number of instances involving acute kidney injury (AKI) and chronic kidney disease (CKD) of unknown etiology (CKDu) have been reported. Additionally, studies have shown a correlation between kidney graft dysfunction in patients with stable kidney transplants after LTPS. These findings indicate that exposure to LTPS may increase the likelihood of kidney transplantation due to the onset of both acute and chronic kidney injuries. Simultaneously, it poses a potential risk to the stability of kidney grafts. Unfortunately, there is limited scientific literature addressing this issue, making it difficult to determine the negative impact that LTPS may have, such as its role as a risk factor for the need of kidney transplantation or as a threat to individuals who have undergone kidney transplants. This study aims to shed light on the immune mechanisms triggered during LTPS infection and their importance in both kidney damage and allograft dysfunction.
Collapse
Affiliation(s)
- Juan Alberto Ruiz-Pacheco
- Investigador por México-CONAHCYT, División de Investigación Quirúrgica, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jalisco, Mexico.
| | | | - Benjamín Gómez-Navarro
- Servicio de Nefrología y trasplantes, Hospital Country 2000, Guadalajara, Jalisco, Mexico
| | - Luis Alberto Castillo-Díaz
- Departamento de Medicina y Ciencias de la Salud, Facultad Interdiciplinaria de Ciencias Biólogicas y de la Salud, Universidad de Sonora, Hermosillo, Mexico
| | - Eliseo Portilla de Buen
- Laboratorio de Investigación quirúrgica, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jalisco, Mexico
| |
Collapse
|
4
|
Lee K, Jang HR, Rabb H. Lymphocytes and innate immune cells in acute kidney injury and repair. Nat Rev Nephrol 2024:10.1038/s41581-024-00875-5. [PMID: 39095505 DOI: 10.1038/s41581-024-00875-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 08/04/2024]
Abstract
Acute kidney injury (AKI) is a common and serious disease entity that affects native kidneys and allografts but for which no specific treatments exist. Complex intrarenal inflammatory processes driven by lymphocytes and innate immune cells have key roles in the development and progression of AKI. Many studies have focused on prevention of early injury in AKI. However, most patients with AKI present after injury is already established. Increasing research is therefore focusing on mechanisms of renal repair following AKI and prevention of progression from AKI to chronic kidney disease. CD4+ and CD8+ T cells, B cells and neutrophils are probably involved in the development and progression of AKI, whereas regulatory T cells, double-negative T cells and type 2 innate lymphoid cells have protective roles. Several immune cells, such as macrophages and natural killer T cells, can have both deleterious and protective effects, depending on their subtype and/or the stage of AKI. The immune system not only participates in injury and repair processes during AKI but also has a role in mediating AKI-induced distant organ dysfunction. Targeted manipulation of immune cells is a promising therapeutic strategy to improve AKI outcomes.
Collapse
Affiliation(s)
- Kyungho Lee
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Cell and Gene Therapy Institute, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Nephrology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hye Ryoun Jang
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Cell and Gene Therapy Institute, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hamid Rabb
- Nephrology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
5
|
Hu X, Wang N, Gao F, Ge S, Lin M, Zhang X, Li T, Li T, Xu C, Huang C, Liang G, Shang W, Xiang F, Feng Y. Prognostic significance of serum Chemerin and neutrophils levels in patients with oral squamous cell carcinoma. Heliyon 2024; 10:e32393. [PMID: 38975159 PMCID: PMC11225754 DOI: 10.1016/j.heliyon.2024.e32393] [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: 01/27/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 07/09/2024] Open
Abstract
Objectives Chemerin, as a novel multifunctional adipokine, is proposed to be involved in high cancer risk and mortality. The present study was aimed to evaluate the prognostic value of serum Chemerin and neutrophils in patients with oral squamous cell carcinoma (OSCC). Materials and methods 120 patients with OSCC were included in this prospective cohort study. The levels of serum Chemerin were measured by enzyme-linked immunosorbent assay (ELISA). We also explored the possible effects of Chemerin on neutrophils' chemokines in OSCC using a real-time PCR, western blotting. Results Levels of serum Chemerin, neutrophils and NLR were significantly higher among non-survivors compared to survivors of OSCC (both P < 0.05). Higher serum Chemerin levels were associated with advanced TNM stage, lymph node metastasis, differentiation and tumor recurrence (both P < 0.05). Serum Chemerin levels correlated with neutrophils and NLR levels (r = 0.708, r = 0.578, both P < 0.05). Based on ROC analysis, Chemerin + NLR predicted OSCC patient mortality with 81.54 % sensitivity and 87.27 % specificity, with an AUC of 0.8898. In a Kaplan-Meier analysis, high serum Chemerin levels, high neutrophil levels and high NLR levels were associated with shorter overall and disease-free survival (both P < 0.05). A univariate and multivariate Cox regression analysis showed that serum Chemerin and neutrophils were independent risk factors for OSCC. (both P < 0.05). QRT-PCR and western blotting results showed that Chemerin upregulated the expression of chemokines IL-17 and CXCL-5 in neutrophils (both P < 0.05). Conclusions Our study suggests that measurement of serum Chemerin and neutrophils might be a useful diagnostic and prognostic biomarker for OSCC patients. Chemerin may promote neutrophils infiltration in OSCC through upregulation of chemokines IL17 and CXCL-5.
Collapse
Affiliation(s)
- Xiaoyuan Hu
- Biological Therapy Center, The Third Affiliated Hospital of Kunming Medical University, Kunzhou Road No. 519, Kunming, Yunnan, 650118, China
| | - Ning Wang
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao, 266003, China
| | - Fei Gao
- Deparment of Pathology, West China Hospital, Sichuan University, Sichuan, 610041, China
| | - Shengyou Ge
- Department of Oral and Maxillofacial Surgery, School of Stomatology and The Affiliated Hospital of Qingdao University, Jiangsu Road No.16, Qingdao, Shandong, 266003, China
| | - Mei Lin
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao, 266003, China
| | - Xuan Zhang
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao, 266003, China
| | - Tongtong Li
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao, 266003, China
| | - Tao Li
- Biological Therapy Center, The Third Affiliated Hospital of Kunming Medical University, Kunzhou Road No. 519, Kunming, Yunnan, 650118, China
| | - Changting Xu
- Biological Therapy Center, The Third Affiliated Hospital of Kunming Medical University, Kunzhou Road No. 519, Kunming, Yunnan, 650118, China
| | - Caixiu Huang
- Biological Therapy Center, The Third Affiliated Hospital of Kunming Medical University, Kunzhou Road No. 519, Kunming, Yunnan, 650118, China
| | - Guicai Liang
- Biological Therapy Center, The Third Affiliated Hospital of Kunming Medical University, Kunzhou Road No. 519, Kunming, Yunnan, 650118, China
| | - Wei Shang
- Department of Oral and Maxillofacial Surgery, School of Stomatology and The Affiliated Hospital of Qingdao University, Jiangsu Road No.16, Qingdao, Shandong, 266003, China
| | - Fenggang Xiang
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao, 266003, China
| | - Yuanyong Feng
- Department of Oral and Maxillofacial Surgery, School of Stomatology and The Affiliated Hospital of Qingdao University, Jiangsu Road No.16, Qingdao, Shandong, 266003, China
| |
Collapse
|
6
|
Lee K, Gharaie S, Kurzhagen JT, Newman-Rivera AM, Arend LJ, Noel S, Rabb H. Double-negative T cells have a reparative role after experimental severe ischemic acute kidney injury. Am J Physiol Renal Physiol 2024; 326:F942-F956. [PMID: 38634135 PMCID: PMC11386976 DOI: 10.1152/ajprenal.00376.2023] [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: 11/27/2023] [Revised: 03/25/2024] [Accepted: 04/15/2024] [Indexed: 04/19/2024] Open
Abstract
T cells mediate organ injury and repair. A proportion of unconventional kidney T cells called double-negative (DN) T cells (TCR+ CD4- CD8-), with anti-inflammatory properties, were previously demonstrated to protect from early injury in moderate experimental acute kidney injury (AKI). However, their role in repair after AKI has not been studied. We hypothesized that DN T cells mediate repair after severe AKI. C57B6 mice underwent severe (40 min) unilateral ischemia-reperfusion injury (IRI). Kidney DN T cells were studied by flow cytometry and compared with gold-standard anti-inflammatory CD4+ regulatory T cells (Tregs). In vitro effects of DN T cells and Tregs on renal tubular epithelial cell (RTEC) repair after injury were quantified with live-cell analysis. DN T cells, Tregs, CD4, or vehicle were adoptively transferred after severe AKI. Glomerular filtration rate (GFR) was measured using fluorescein isothiocyanate (FITC)-sinistrin. Fibrosis was assessed with Masson's trichrome staining. Profibrotic genes were measured with qRT-PCR. Percentages and the numbers of DN T cells substantially decreased during repair phase after severe AKI, as well as their activation and proliferation. Both DN T cells and Tregs accelerated RTEC cell repair in vitro. Post-AKI transfer of DN T cells reduced kidney fibrosis and improved GFR, as did Treg transfer. DN T cell transfer lowered transforming growth factor (TGF)β1 and α-smooth muscle actin (αSMA) expression. DN T cells reduced effector-memory CD4+ T cells and IL-17 expression. DN T cells undergo quantitative and phenotypical changes after severe AKI, accelerate RTEC repair in vitro as well as improve GFR and renal fibrosis in vivo. DN T cells have potential as immunotherapy to accelerate repair after AKI.NEW & NOTEWORTHY Double-negative (DN) T cells (CD4- CD8-) are unconventional kidney T cells with regulatory abilities. Their role in repair from acute kidney injury (AKI) is unknown. Kidney DN T cell population decreased during repair after ischemic AKI, in contrast to regulatory T cells (Tregs) which increased. DN T cell administration accelerated tubular repair in vitro, while after severe in vivo ischemic injury reduced kidney fibrosis and increased glomerular filtration rate (GFR). DN T cell infusion is a potential therapeutic agent to improve outcome from severe AKI.
Collapse
Affiliation(s)
- Kyungho Lee
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Nephrology, Department of Medicine, Cell and Gene Therapy Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sepideh Gharaie
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Johanna T Kurzhagen
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Andrea M Newman-Rivera
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Lois J Arend
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Sanjeev Noel
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Hamid Rabb
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
7
|
Liu H, Xiang W, Wu W, Zhou G, Yuan J. Associations of systemic inflammatory regulators with CKD and kidney function: evidence from the bidirectional mendelian randomization study. BMC Nephrol 2024; 25:161. [PMID: 38730296 PMCID: PMC11088104 DOI: 10.1186/s12882-024-03590-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 04/26/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Previous observational studies have reported that systemic inflammatory regulators are related to the development of chronic kidney disease (CKD); however, whether these associations are causal remains unclear. The current study aimed to investigate the potential causal relationships between systemic inflammatory regulators and CKD and kidney function. METHOD We performed bidirectional two-sample Mendelian randomization (MR) analyses to infer the underlying causal associations between 41 systemic inflammatory regulators and CKD and kidney function. The inverse-variance weighting (IVW) test was used as the primary analysis method. In addition, sensitivity analyses were executed via the Mendelian randomization pleiotropy residual sum and outlier (MR-PRESSO) test and the weighted median test. RESULTS The findings revealed 12 suggestive associations between 11 genetically predicted systemic inflammatory regulators and CKD or kidney function in the forward analyses, including 4 for CKD, 3 for blood urea nitrogen (BUN), 4 for eGFRcrea and 1 for eGFRcys. In the other direction, we identified 6 significant causal associations, including CKD with granulocyte-colony stimulating factor (GCSF) (IVW β = 0.145; 95% CI, 0.042 to 0.248; P = 0.006), CKD with stem cell factor (SCF) (IVW β = 0.228; 95% CI, 0.133 to 0.323; P = 2.40 × 10- 6), eGFRcrea with SCF (IVW β =-2.90; 95% CI, -3.934 to -1.867; P = 3.76 × 10- 8), eGFRcys with GCSF (IVW β =-1.382; 95% CI, -2.404 to -0.361; P = 0.008), eGFRcys with interferon gamma (IFNg) (IVW β =-1.339; 95% CI, -2.313 to -0.366; P = 0.007) and eGFRcys with vascular endothelial growth factor (VEGF) (IVW β =-1.709; 95% CI, -2.720 to -0.699; P = 9.13 × 10- 4). CONCLUSIONS Our findings support causal links between systemic inflammatory regulators and CKD or kidney function both in the forward and reverse MR analyses.
Collapse
Affiliation(s)
- Hailang Liu
- Department of Urology, Wuhan Integrated TCM & Western Medicine Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan No.1 Hospital, Wuhan, China
| | - Wei Xiang
- Department of Urology, Wuhan Integrated TCM & Western Medicine Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan No.1 Hospital, Wuhan, China
| | - Wei Wu
- Department of Urology, Wuhan Integrated TCM & Western Medicine Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan No.1 Hospital, Wuhan, China
| | - Gaofeng Zhou
- Department of Urology, Wuhan Integrated TCM & Western Medicine Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan No.1 Hospital, Wuhan, China.
| | - Jingdong Yuan
- Department of Urology, Wuhan Integrated TCM & Western Medicine Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan No.1 Hospital, Wuhan, China.
| |
Collapse
|
8
|
Schmauch E, Piening B, Mohebnasab M, Xia B, Zhu C, Stern J, Zhang W, Dowdell AK, Kim JI, Andrijevic D, Khalil K, Jaffe IS, Loza BL, Gragert L, Camellato BR, Oliveira MF, O'Brien DP, Chen HM, Weldon E, Gao H, Gandla D, Chang A, Bhatt R, Gao S, Lin X, Reddy KP, Kagermazova L, Habara AH, Widawsky S, Liang FX, Sall J, Loupy A, Heguy A, Taylor SEB, Zhu Y, Michael B, Jiang L, Jian R, Chong AS, Fairchild RL, Linna-Kuosmanen S, Kaikkonen MU, Tatapudi V, Lorber M, Ayares D, Mangiola M, Narula N, Moazami N, Pass H, Herati RS, Griesemer A, Kellis M, Snyder MP, Montgomery RA, Boeke JD, Keating BJ. Integrative multi-omics profiling in human decedents receiving pig heart xenografts. Nat Med 2024; 30:1448-1460. [PMID: 38760586 DOI: 10.1038/s41591-024-02972-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 04/03/2024] [Indexed: 05/19/2024]
Abstract
In a previous study, heart xenografts from 10-gene-edited pigs transplanted into two human decedents did not show evidence of acute-onset cellular- or antibody-mediated rejection. Here, to better understand the detailed molecular landscape following xenotransplantation, we carried out bulk and single-cell transcriptomics, lipidomics, proteomics and metabolomics on blood samples obtained from the transplanted decedents every 6 h, as well as histological and transcriptomic tissue profiling. We observed substantial early immune responses in peripheral blood mononuclear cells and xenograft tissue obtained from decedent 1 (male), associated with downstream T cell and natural killer cell activity. Longitudinal analyses indicated the presence of ischemia reperfusion injury, exacerbated by inadequate immunosuppression of T cells, consistent with previous findings of perioperative cardiac xenograft dysfunction in pig-to-nonhuman primate studies. Moreover, at 42 h after transplantation, substantial alterations in cellular metabolism and liver-damage pathways occurred, correlating with profound organ-wide physiological dysfunction. By contrast, relatively minor changes in RNA, protein, lipid and metabolism profiles were observed in decedent 2 (female) as compared to decedent 1. Overall, these multi-omics analyses delineate distinct responses to cardiac xenotransplantation in the two human decedents and reveal new insights into early molecular and immune responses after xenotransplantation. These findings may aid in the development of targeted therapeutic approaches to limit ischemia reperfusion injury-related phenotypes and improve outcomes.
Collapse
Affiliation(s)
- Eloi Schmauch
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
| | - Brian Piening
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR, USA
| | - Maedeh Mohebnasab
- Division of Molecular Genetics Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Bo Xia
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA
- Society of Fellows, Harvard University, Cambridge, MA, USA
| | - Chenchen Zhu
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Jeffrey Stern
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Weimin Zhang
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA
| | - Alexa K Dowdell
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR, USA
| | - Jacqueline I Kim
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - David Andrijevic
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Karen Khalil
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
| | - Ian S Jaffe
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Bao-Li Loza
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Loren Gragert
- Division of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | | | | | | | - Han M Chen
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Elaina Weldon
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Hui Gao
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Divya Gandla
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew Chang
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Riyana Bhatt
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah Gao
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiangping Lin
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Kriyana P Reddy
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Alawi H Habara
- Department of Biochemistry, College of Medicine, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Sophie Widawsky
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Feng-Xia Liang
- DART Microscopy Laboratory, NYU Langone Health, New York, NY, USA
| | - Joseph Sall
- DART Microscopy Laboratory, NYU Langone Health, New York, NY, USA
| | - Alexandre Loupy
- Université Paris Cité, Paris Institute for Transplantation and Organ Regeneration, Paris, France
| | - Adriana Heguy
- Genome Technology Center, NYU Langone Health, New York, NY, USA
| | | | - Yinan Zhu
- Division of Molecular Genetics Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Basil Michael
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Lihua Jiang
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Ruiqi Jian
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Anita S Chong
- Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Robert L Fairchild
- Department of Inflammation and Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Suvi Linna-Kuosmanen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Minna U Kaikkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Vasishta Tatapudi
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | | | | | - Massimo Mangiola
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
| | - Navneet Narula
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Nader Moazami
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Cardiothoracic Surgery, NYU Langone Health, New York, NY, USA
| | - Harvey Pass
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Cardiothoracic Surgery, NYU Langone Health, New York, NY, USA
| | - Ramin S Herati
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Adam Griesemer
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Manolis Kellis
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
| | | | - Robert A Montgomery
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Jef D Boeke
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
- Department of Biomedical Engineering, NYU Tandon School of Engineering, Brooklyn, NY, USA
| | - Brendan J Keating
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA.
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA.
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
9
|
Ullah MM, Collett JA, Monroe JC, Traktuev D, Coleman M, March KL, Basile DP. Subcutaneous injection of adipose stromal cell-secretome improves renal function and reduces inflammation in established acute kidney injury. Stem Cell Res Ther 2024; 15:119. [PMID: 38659070 PMCID: PMC11040889 DOI: 10.1186/s13287-024-03736-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: 08/14/2023] [Accepted: 04/17/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Adipose stromal cells (ASC) are a form of mesenchymal stromal cells that elicit effects primarily via secreted factors, which may have advantages for the treatment of injury or disease. Several previous studies have demonstrated a protective role for MSC/ASC on mitigating acute kidney injury but whether ASC derived factors could hasten recovery from established injury has not been evaluated. METHODS We generated a concentrated secretome (CS) of human ASC under well-defined conditions and evaluated its ability to improve the recovery of renal function in a preclinical model of acute kidney injury (AKI) in rats. 24 h following bilateral ischemia/reperfusion (I/R), rats were randomized following determination of plasma creatinine into groups receiving vehicle -control or ASC-CS treatment by subcutaneous injection (2 mg protein/kg) and monitored for evaluation of renal function, structure and inflammation. RESULTS Renal function, assessed by plasma creatinine levels, recovered faster in ASC-CS treated rats vs vehicle. The most prominent difference between the ASC-CS treated vs vehicle was observed in rats with the most severe degree of initial injury (Pcr > 3.0 mg/dl 24 h post I/R), whereas rats with less severe injury (Pcr < 2.9 mg/dl) recovered quickly regardless of treatment. The quicker recovery of ASC-treated rats with severe injury was associated with less tissue damage, inflammation, and lower plasma angiopoietin 2. In vitro, ASC-CS attenuated the activation of the Th17 phenotype in lymphocytes isolated from injured kidneys. CONCLUSIONS Taken together, these data suggest that ASC-CS represents a potent therapeutic option to improve established AKI.
Collapse
Affiliation(s)
- Md Mahbub Ullah
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, 635 Barnhill Dr. MS 2063, Indianapolis, IN, 46202, USA
| | - Jason A Collett
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, 635 Barnhill Dr. MS 2063, Indianapolis, IN, 46202, USA
| | - Jacob C Monroe
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, 635 Barnhill Dr. MS 2063, Indianapolis, IN, 46202, USA
| | - Dmitry Traktuev
- Division of Cardiovascular Medicine and Center for Regenerative Medicine, University of Florida, Gainesville, FL, USA
- Theratome Bio, Inc., Indianapolis, IN, USA
| | - Michael Coleman
- Division of Cardiovascular Medicine and Center for Regenerative Medicine, University of Florida, Gainesville, FL, USA
| | - Keith L March
- Division of Cardiovascular Medicine and Center for Regenerative Medicine, University of Florida, Gainesville, FL, USA
- Theratome Bio, Inc., Indianapolis, IN, USA
| | - David P Basile
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, 635 Barnhill Dr. MS 2063, Indianapolis, IN, 46202, USA.
| |
Collapse
|
10
|
Zhang T, Huo H, Zhang Y, Tao J, Yang J, Rong X, Yang Y. Th17 cells: A new target in kidney disease research. Int Rev Immunol 2024; 43:263-279. [PMID: 38439681 DOI: 10.1080/08830185.2024.2321901] [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: 08/28/2023] [Revised: 12/26/2023] [Accepted: 01/09/2024] [Indexed: 03/06/2024]
Abstract
Type 17 T helper (Th17) cells, which are a subtype of CD4+ T helper cells, secrete pro-inflammatory cytokines such as IL-17A, IL-17F, IL-21, IL-22, and GM-CSF, which play crucial roles in immune defence and protection against fungal and extracellular pathogen invasion. However, dysfunction of Th17 cell immunity mediates inflammatory responses and exacerbates tissue damage. This pathological process initiated by Th17 cells is common in kidney diseases associated with renal injury, such as glomerulonephritis, lupus nephritis, IgA nephropathy, hypertensive nephropathy, diabetic kidney disease and acute kidney injury. Therefore, targeting Th17 cells to treat kidney diseases has been a hot topic in recent years. This article reviews the mechanisms of Th17 cell-mediated inflammation and autoimmune responses in kidney diseases and discusses the related clinical drugs that modulate Th17 cell fate in kidney disease treatment.
Collapse
Affiliation(s)
- Tao Zhang
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hongyan Huo
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yinghui Zhang
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jie Tao
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Junzheng Yang
- Guangdong Nephrotic Drug Engineering Technology Research Center, The R&D Center of Drug for Renal Diseases, Consun Pharmaceutical Group, Guangzhou, Guangdong, China
| | - Xianglu Rong
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yiqi Yang
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
11
|
Wang Q, Zhen W, Lippi G, Liu Q. The effect of Astragali Radix-Radix Angelica Sinensis on acute kidney injury: a network pharmacology and molecular docking study. Transl Androl Urol 2024; 13:91-103. [PMID: 38404557 PMCID: PMC10891378 DOI: 10.21037/tau-23-562] [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: 11/04/2023] [Accepted: 01/10/2024] [Indexed: 02/27/2024] Open
Abstract
Background Acute kidney injury (AKI) is a devastating clinical syndrome with high mortality rate attributed to lack of effective treatment. The herbal pair of Astragali Radix (AR) and Radix Angelica Sinensis (RAS) is a commonly prescribed herbal formula or is added to other traditional Chinese medicine (TCM) prescriptions for the treatment of kidney diseases. AR-RAS has certain protective effects on AKI in experiments, but the relevant mechanisms have yet to be clear. So this study aims to explore the mechanism of action of AR-RAS in AKI by combining network pharmacology and molecular docking methods. Methods In Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP), the major AR-RAS chemical components and associated action targets were found and screened. The DrugBank and GeneCards databases were used to find AKI-related targets. The targets that are in close relationship with AKI were obtained from Therapeutic Target database (TTD), Online Mendelian Inheritance in Man (OMIM), and PharmGKB databases. The "herb-active ingredient-target" network was drawn by Cytoscape 3.8.0 software. The Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database was used to build the protein-protein interaction network. Bioconductor/R was used to examine Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment. AR-RAS components and critical targets were docked using the AutoDock Vina program. Results A compound-target network, built by screening and analyzing the results, allowed to identify 19 active components and 101 possible therapeutic targets for AKI. The main ingredients were quercetin, kaempferol, 7-o-methylisocronulatol, formononetin and isorhamnetin. The key targets included AKT serine/threonine kinase 1 (AKT1), nuclear receptor coactivator 1 (NCOA1), JUN, estrogen receptor alpha (ESR1) and mitogen-activated protein kinase 8 (MAPK8). These molecules are targeted by pathways such as the calcium signaling route, the tumor necrosis factor (TNF) signaling pathway and the interleukin-17 (IL-17) signaling pathway, as well as the development of T helper 17 cells. Molecular docking demonstrated that AR-active RAS components exhibited strong binding activities to probable targets of AKI. Conclusions We described here the potential active ingredients, possible targets responsible for the efficacy of AR-RAS in AKI treatment, providing a theoretical basis for further research.
Collapse
Affiliation(s)
- Qin Wang
- Suzhou Medical College of Soochow University, Suzhou, China
- Department of Nephrology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Wenrui Zhen
- Department of Intervention Therapy, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Giuseppe Lippi
- Section of Clinical Biochemistry and School of Medicine, University of Verona, Verona, Italy
| | - Qi Liu
- Suzhou Medical College of Soochow University, Suzhou, China
- Department of Nephrology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
12
|
Yang L, He T, Yu Y. Uric acid promotes interleukin-17 expression to cause kidney injury. J Biochem Mol Toxicol 2024; 38:e23550. [PMID: 37815028 DOI: 10.1002/jbt.23550] [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: 02/06/2023] [Revised: 08/02/2023] [Accepted: 09/27/2023] [Indexed: 10/11/2023]
Abstract
Uric acid, an oxidation end-product of purine metabolism, is reportedly to be a risk factor for kidney injury. However, its underlying mechanism is still a mystery. This study aimed to reveal the detailed roles of uric acid in inducing kidney injury and the possible mechanisms. Injection of rats with uric acid significantly increased tubular injury score, and levels of blood urea nitrogen, serum creatinine, and urine kidney injury molecule-1. Uric acid increased the expression of collagen I, alpha-smooth muscle actin, tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6. Kyoto Encyclopedia of Genes and Genomes analysis result showed the IL-17 signaling pathway as the most significantly enriched pathway involved in hyperuricemia-related kidney injury. Long-term injection of uric acid induced significant production of IL-17 and recruitment of Th17 cells. Treating rats with the anti-IL-17 mAb attenuated uric acid-induced kidney injury, accompanied by the inactivation of nuclear factor-κB (NF-κB). In conclusion, uric acid was confirmed to be a risk factor for kidney injury via inducing IL-17 expression. Neutralization of IL-17 using the specific mAb relieved uric acid-induced kidney injury via inhibition of NF-κB signaling.
Collapse
Affiliation(s)
- Lina Yang
- Department of Nephrology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, PR China
| | - Tianwei He
- Department of Nephrology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, PR China
| | - Yanming Yu
- Department of Nephrology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, PR China
| |
Collapse
|
13
|
Ullah MM, Collett JA, Bacallao RL, Basile DP. Impaired hemodynamic renal reserve response following recovery from established acute kidney injury and improvement by hydrodynamic isotonic fluid delivery. Am J Physiol Renal Physiol 2024; 326:F86-F94. [PMID: 37881874 PMCID: PMC11194053 DOI: 10.1152/ajprenal.00204.2023] [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: 07/18/2023] [Revised: 10/04/2023] [Accepted: 10/18/2023] [Indexed: 10/27/2023] Open
Abstract
Renal reserve capacity may be compromised following recovery from acute kidney injury (AKI) and could be used to identify impaired renal function in the face of restored glomerular filtration rate (GFR) or plasma creatinine. To investigate the loss of hemodynamic renal reserve responses following recovery in a model of AKI, rats were subjected to left unilateral renal ischemia-reperfusion (I/R) injury and contralateral nephrectomy and allowed to recover for 5 wk. Some rats were treated 24 h post-I/R by hydrodynamic isotonic fluid delivery (AKI-HIFD) of saline through the renal vein, previously shown to improve recovery and inflammation relative to control rats that received saline through the vena cava (AKI-VC). At 5 wk after surgery, plasma creatinine and GFR recovered to levels observed in uninephrectomized sham controls. Baseline renal blood flow (RBF) was not different between AKI or sham groups, but infusion of l-arginine (7.5 mg/kg/min) significantly increased RBF in sham controls, whereas the RBF response to l-arginine was significantly reduced in AKI-VC rats relative to sham rats (22.6 ± 2.2% vs. 13.8 ± 1.8%, P < 0.05). RBF responses were partially protected in AKI-HIFD rats relative to AKI-VC rats (17.0 ± 2.2%) and were not significantly different from sham rats. Capillary rarefaction observed in AKI-VC rats was significantly protected in AKI-HIFD rats. There was also a significant increase in T helper 17 cell infiltration and interstitial fibrosis in AKI-VC rats versus sham rats, which was not present in AKI-HIFD rats. These data suggest that recovery from AKI results in impaired hemodynamic reserve and that associated CKD progression may be mitigated by HIFD in the early post-AKI period.NEW & NOTEWORTHY Despite the apparent recovery of renal filtration function following acute kidney injury (AKI) in rats, the renal hemodynamic reserve response is significantly attenuated, suggesting that clinical evaluation of this parameter may provide information on the potential development of chronic kidney disease. Treatments such as hydrodynamic isotonic fluid delivery, or other treatments in the early post-AKI period, could minimize chronic inflammation or loss of microvessels with the potential to promote a more favorable outcome on long-term function.
Collapse
Affiliation(s)
- Md Mahbub Ullah
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Jason A Collett
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Robert L Bacallao
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, United States
| | - David P Basile
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| |
Collapse
|
14
|
Zhu Q, Ren S, Sun Z, Qin J, Sheng X. Identification of biomarkers of renal ischemia-reperfusion injury by bioinformatics analysis and single-cell sequencing analysis combined with in vivo validation. Transpl Immunol 2023; 81:101928. [PMID: 37704087 DOI: 10.1016/j.trim.2023.101928] [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: 06/14/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND Renal ischemia-reperfusion injury (IRI) is a serious clinical complication of kidney injury. This research dealt with investigating the hub genes and pathways associated with renal IRI. METHODS The transcriptome expression dataset of mouse renal ischemia samples (GSE39548) was obtained from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) were filtered by R software for key genes utilized for gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, and gene enrichment analysis (GSEA). The gene co-expression network was developed by WGCNA analysis to screen important modules. Hub genes from the intersection of DEGs and WGCNA were subjected to protein-protein interaction (PPI) network. The biomarkers obtained by SVM-REF and LASSO algorithm were validated by other datasets and subjected to GSEA analysis. The expression of biomarkers in renal IRI was detected by qRT-PCR and subjected to single-cell analysis. RESULTS A total of 157 DEGs were discovered. Biological function analysis depicted that the DEGs were primarily involved in cytokine-cytokine receptor interaction, as well as the signaling pathways IL-17, MAPK, and TNF. The intersection of DEGs and the genes obtained by WGCNA analysis yielded 149 hubs genes. Based on SVM-REF and LASSO algorithm, cyp1a1 and pdk4 were determined as potential biomarkers in individuals with renal ischemia and showed good diagnostic value. qRT-PCR results depicted that cyp1a1 and pdk4 were significantly up-regulated in renal ischemia mice (P < 0.05). Finally, the single-cell analysis identified the expression of Cyp1a1 and Pdk4 in mice kidney tissue. CONCLUSION cyp1a1 and pdk4 were identified to play important roles in renal IRI. This research provides a new perspective and basis for studying the pathogenesis of renal IRI and developing new treatments.
Collapse
Affiliation(s)
- Qin Zhu
- Department of Hand Surgery, Nantong University Affiliated Hospital, Nantong 226001, China
| | - Shiqi Ren
- Department of Hand Surgery, Nantong University Affiliated Hospital, Nantong 226001, China
| | - Zhaoyang Sun
- Department of Hand Surgery, Nantong University Affiliated Hospital, Nantong 226001, China
| | - Jun Qin
- Department of Trauma Center, Affiliated Hospital of Nantong University, Nantong 226001, China.
| | - Xiaoming Sheng
- Department of Trauma Center, Affiliated Hospital of Nantong University, Nantong 226001, China.
| |
Collapse
|
15
|
Cartwright BM, Corso JN, Lightner J, Whitted C, Torrenegra RD, Krishnan K, Palau VE. Achyrocline B (3,5 dihydroxy-6,7,8-trimethoxyflavone) synergizes with 5-fluorouracil allowing for dose reduction and reduced off-target toxicity in the treatment of colonic and pancreatic cancers. Biomed Pharmacother 2023; 167:115546. [PMID: 37741250 DOI: 10.1016/j.biopha.2023.115546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/12/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023] Open
Abstract
Surgically unresectable colorectal and pancreatic carcinomas have a high rate of mortality as current therapeutic options are limited. One common chemotherapeutic used to broadly treat both cancers is 5-flurouracil (5-Fu); however, treatment serves only to slow progression of the disease and comes with many side effects due to 5-Fu's intrinsic toxicity. Thus, strategies to decrease the dose of 5-Fu utilized therapeutically as well as reduce 5-Fu's off-target toxicity are paramount. Using cell models of colorectal and pancreatic cancers, we show that cotreatment with Achyrocline B (3,5 dihydroxy-6,7,8-trimethoxyflavone, AcB), a natural flavone from Achyrocline bogotensis, allows for four-fold reduction in 5-Fu dosage without loss of efficacy. We further show that the action of AcB is due to continued cell cycle progression despite 5-Fu pressure to synchronize at the G1/S threshold. In addition to AcB's effect on cancer cells, we found that AcB can directly reduce toxicity of 5-Fu in cells mimicking non-cancerous tissues. These in vitro results are then supported by xenograft modeling. AcB was shown to increase apoptosis in tumors leading to degeneration of the outer tumoral boundary. Furthermore, in 5-Fu treated animals it was found that AcB provided protection to the intestinal tract as indicated by preserved histological and immunohistochemical features. These results show promise for a new adjuvant therapy for colorectal and pancreatic carcinomas that not only reduces tumor progression, but more importantly has the potential to improve patient quality of life.
Collapse
Affiliation(s)
- Brian M Cartwright
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, 37614, United States; Department of Pathology, ETSU Quillen College of Medicine, Johnson City, TN, 37614, United States
| | - Jaclyn N Corso
- Department of Internal Medicine, ETSU Quillen College of Medicine, Johnson City, TN, 37614, United States
| | - Janet Lightner
- Department of Internal Medicine, ETSU Quillen College of Medicine, Johnson City, TN, 37614, United States
| | - Crystal Whitted
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, 37614, United States
| | - Ruben D Torrenegra
- Productos Naturales, Universidad de Ciencias Aplicadas y Ambientales, Bogota, Colombia
| | - Koyamangalath Krishnan
- Department of Internal Medicine, ETSU Quillen College of Medicine, Johnson City, TN, 37614, United States
| | - Victoria E Palau
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, 37614, United States; Productos Naturales, Universidad de Ciencias Aplicadas y Ambientales, Bogota, Colombia.
| |
Collapse
|
16
|
Guo Y, Cen K, Hong K, Mai Y, Jiang M. Construction of a neural network diagnostic model for renal fibrosis and investigation of immune infiltration characteristics. Front Immunol 2023; 14:1183088. [PMID: 37359552 PMCID: PMC10288286 DOI: 10.3389/fimmu.2023.1183088] [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: 03/09/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
Background Recently, the incidence rate of renal fibrosis has been increasing worldwide, greatly increasing the burden on society. However, the diagnostic and therapeutic tools available for the disease are insufficient, necessitating the screening of potential biomarkers to predict renal fibrosis. Methods Using the Gene Expression Omnibus (GEO) database, we obtained two gene array datasets (GSE76882 and GSE22459) from patients with renal fibrosis and healthy individuals. We identified differentially expressed genes (DEGs) between renal fibrosis and normal tissues and analyzed possible diagnostic biomarkers using machine learning. The diagnostic effect of the candidate markers was evaluated using receiver operating characteristic (ROC) curves and verified their expression using Reverse transcription quantitative polymerase chain reaction (RT-qPCR). The CIBERSORT algorithm was used to determine the proportions of 22 types of immune cells in patients with renal fibrosis, and the correlation between biomarker expression and the proportion of immune cells was studied. Finally, we developed an artificial neural network model of renal fibrosis. Results Four candidate genes namely DOCK2, SLC1A3, SOX9 and TARP were identified as biomarkers of renal fibrosis, with the area under the ROC curve (AUC) values higher than 0.75. Next, we verified the expression of these genes by RT-qPCR. Subsequently, we revealed the potential disorder of immune cells in the renal fibrosis group through CIBERSORT analysis and found that immune cells were highly correlated with the expression of candidate markers. Conclusion DOCK2, SLC1A3, SOX9, and TARP were identified as potential diagnostic genes for renal fibrosis, and the most relevant immune cells were identified. Our findings provide potential biomarkers for the diagnosis of renal fibrosis.
Collapse
Affiliation(s)
- Yangyang Guo
- Department of General Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Department of Urology Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Kenan Cen
- Department of General Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Kai Hong
- Department of General Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yifeng Mai
- Department of General Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Minghui Jiang
- Department of Urology Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| |
Collapse
|
17
|
Wang L, Peng F, Li ZH, Deng YF, Ruan MN, Mao ZG, Li L. Identification of AKI signatures and classification patterns in ccRCC based on machine learning. Front Med (Lausanne) 2023; 10:1195678. [PMID: 37293297 PMCID: PMC10244623 DOI: 10.3389/fmed.2023.1195678] [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: 03/28/2023] [Accepted: 05/03/2023] [Indexed: 06/10/2023] Open
Abstract
Background Acute kidney injury can be mitigated if detected early. There are limited biomarkers for predicting acute kidney injury (AKI). In this study, we used public databases with machine learning algorithms to identify novel biomarkers to predict AKI. In addition, the interaction between AKI and clear cell renal cell carcinoma (ccRCC) remain elusive. Methods Four public AKI datasets (GSE126805, GSE139061, GSE30718, and GSE90861) treated as discovery datasets and one (GSE43974) treated as a validation dataset were downloaded from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) between AKI and normal kidney tissues were identified using the R package limma. Four machine learning algorithms were used to identify the novel AKI biomarkers. The correlations between the seven biomarkers and immune cells or their components were calculated using the R package ggcor. Furthermore, two distinct ccRCC subtypes with different prognoses and immune characteristics were identified and verified using seven novel biomarkers. Results Seven robust AKI signatures were identified using the four machine learning methods. The immune infiltration analysis revealed that the numbers of activated CD4 T cells, CD56dim natural killer cells, eosinophils, mast cells, memory B cells, natural killer T cells, neutrophils, T follicular helper cells, and type 1 T helper cells were significantly higher in the AKI cluster. The nomogram for prediction of AKI risk demonstrated satisfactory discrimination with an Area Under the Curve (AUC) of 0.919 in the training set and 0.945 in the testing set. In addition, the calibration plot demonstrated few errors between the predicted and actual values. In a separate analysis, the immune components and cellular differences between the two ccRCC subtypes based on their AKI signatures were compared. Patients in the CS1 had better overall survival, progression-free survival, drug sensitivity, and survival probability. Conclusion Our study identified seven distinct AKI-related biomarkers based on four machine learning methods and proposed a nomogram for stratified AKI risk prediction. We also confirmed that AKI signatures were valuable for predicting ccRCC prognosis. The current work not only sheds light on the early prediction of AKI, but also provides new insights into the correlation between AKI and ccRCC.
Collapse
Affiliation(s)
- Li Wang
- Department of Nephrology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Fei Peng
- Department of Cardiology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Zhen Hua Li
- Department of Cardiology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yu Fei Deng
- Department of Nephrology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Meng Na Ruan
- Department of Nephrology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhi Guo Mao
- Department of Nephrology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Lin Li
- Department of Nephrology, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
18
|
Kurzhagen JT, Noel S, Lee K, Sadasivam M, Gharaie S, Ankireddy A, Lee SA, Newman-Rivera A, Gong J, Arend LJ, Hamad AR, Reddy SP, Rabb H. T Cell Nrf2/Keap1 Gene Editing Using CRISPR/Cas9 and Experimental Kidney Ischemia-Reperfusion Injury. Antioxid Redox Signal 2023; 38:959-973. [PMID: 36734409 PMCID: PMC10171956 DOI: 10.1089/ars.2022.0058] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 12/21/2022] [Accepted: 01/05/2023] [Indexed: 02/04/2023]
Abstract
Aims: T cells play pathophysiologic roles in kidney ischemia-reperfusion injury (IRI), and the nuclear factor erythroid 2-related factor 2/kelch-like ECH-associated protein 1 (Nrf2/Keap1) pathway regulates T cell responses. We hypothesized that clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9)-mediated Keap1-knockout (KO) augments Nrf2 antioxidant potential of CD4+ T cells, and that Keap1-KO CD4+ T cell immunotherapy protects from kidney IRI. Results: CD4+ T cell Keap1-KO resulted in significant increase of Nrf2 target genes NAD(P)H quinone dehydrogenase 1, heme oxygenase 1, glutamate-cysteine ligase catalytic subunit, and glutamate-cysteine ligase modifier subunit. Keap1-KO cells displayed no signs of exhaustion, and had significantly lower levels of interleukin 2 (IL2) and IL6 in normoxic conditions, but increased interferon gamma in hypoxic conditions in vitro. In vivo, adoptive transfer of Keap1-KO CD4+ T cells before IRI improved kidney function in T cell-deficient nu/nu mice compared with mice receiving unedited control CD4+ T cells. Keap1-KO CD4+ T cells isolated from recipient kidneys 24 h post IR were less activated compared with unedited CD4+ T cells, isolated from control kidneys. Innovation: Editing Nrf2/Keap1 pathway in murine T cells using CRISPR/Cas9 is an innovative and promising immunotherapy approach for kidney IRI and possibly other solid organ IRI. Conclusion: CRISPR/Cas9-mediated Keap1-KO increased Nrf2-regulated antioxidant gene expression in murine CD4+ T cells, modified responses to in vitro hypoxia and in vivo kidney IRI. Gene editing targeting the Nrf2/Keap1 pathway in T cells is a promising approach for immune-mediated kidney diseases.
Collapse
Affiliation(s)
- Johanna T. Kurzhagen
- Division of Nephrology and Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sanjeev Noel
- Division of Nephrology and Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kyungho Lee
- Division of Nephrology and Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mohanraj Sadasivam
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sepideh Gharaie
- Division of Nephrology and Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aparna Ankireddy
- Department of Pediatrics, University of Illinois, Chicago, Illinois, USA
| | - Sul A. Lee
- Division of Nephrology and Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrea Newman-Rivera
- Division of Nephrology and Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jing Gong
- Division of Nephrology and Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lois J. Arend
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Abdel R.A. Hamad
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sekhar P. Reddy
- Department of Pediatrics, University of Illinois, Chicago, Illinois, USA
- Department of Pathology, and University of Illinois, Chicago, Illinois, USA
- University of Illinois Cancer Center, University of Illinois, Chicago, Illinois, USA
| | - Hamid Rabb
- Division of Nephrology and Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
19
|
Goel H, Printz RL, Shiota C, Estes SK, Pannala V, AbdulHameed MDM, Shiota M, Wallqvist A. Assessing Kidney Injury Induced by Mercuric Chloride in Guinea Pigs with In Vivo and In Vitro Experiments. Int J Mol Sci 2023; 24:7434. [PMID: 37108594 PMCID: PMC10138559 DOI: 10.3390/ijms24087434] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/04/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Acute kidney injury, which is associated with high levels of morbidity and mortality, affects a significant number of individuals, and can be triggered by multiple factors, such as medications, exposure to toxic chemicals or other substances, disease, and trauma. Because the kidney is a critical organ, understanding and identifying early cellular or gene-level changes can provide a foundation for designing medical interventions. In our earlier work, we identified gene modules anchored to histopathology phenotypes associated with toxicant-induced liver and kidney injuries. Here, using in vivo and in vitro experiments, we assessed and validated these kidney injury-associated modules by analyzing gene expression data from the kidneys of male Hartley guinea pigs exposed to mercuric chloride. Using plasma creatinine levels and cell-viability assays as measures of the extent of renal dysfunction under in vivo and in vitro conditions, we performed an initial range-finding study to identify the appropriate doses and exposure times associated with mild and severe kidney injuries. We then monitored changes in kidney gene expression at the selected doses and time points post-toxicant exposure to characterize the mechanisms of kidney injury. Our injury module-based analysis revealed a dose-dependent activation of several phenotypic cellular processes associated with dilatation, necrosis, and fibrogenesis that were common across the experimental platforms and indicative of processes that initiate kidney damage. Furthermore, a comparison of activated injury modules between guinea pigs and rats indicated a strong correlation between the modules, highlighting their potential for cross-species translational studies.
Collapse
Affiliation(s)
- Himanshu Goel
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Frederick, MD 21702, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Richard L. Printz
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Chiyo Shiota
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Shanea K. Estes
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Venkat Pannala
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Frederick, MD 21702, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Mohamed Diwan M. AbdulHameed
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Frederick, MD 21702, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Masakazu Shiota
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Frederick, MD 21702, USA
| |
Collapse
|
20
|
Gal-9/Tim-3 signaling pathway activation suppresses the generation of Th17 cells and promotes the induction of Foxp3 + regulatory T cells in renal ischemia-reperfusion injury. Mol Immunol 2023; 156:136-147. [PMID: 36921488 DOI: 10.1016/j.molimm.2023.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/03/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023]
Abstract
CD4+ T cells mediate the pathogenesis of renal ischemia-reperfusion injury (IRI). Emerging research suggests that a Th17/regulatory T cell (Treg) imbalance plays a pivotal role in the development of renal IRI. A recently identified negative checkpoint protein, T cell immunoglobulin domain and mucin domain family 3 (Tim-3), inhibits the immune response by binding to its ligand, galectin-9 (Gal-9). However, the role of the Gal-9/Tim-3 signaling pathway in the regulation of CD4+ T cell subsets in renal IRI remains unclear. In this study, we investigated the effect of the Gal-9/Tim-3 signaling pathway on Th17/Treg subsets in renal IRI using a mouse model. Renal IRI induced the expression of Gal-9 in renal tubular epithelial cells and increased the proportion of Tim-3+ Th17 cells and Tim-3+ forkhead box P3 (Foxp3)+ Treg cells in the ischemia-reperfusion (IR) kidneys. Administration of rAAV9-Gal-9 suppressed kidney inflammation, reduced the mortality of mice with renal IRI, increased Foxp3+ Treg cells, and reduced Th17 cells. In contrast, the blockade of Tim-3 in vivo using an anti-Tim-3 monoclonal antibody aggravated renal inflammation, decreased Foxp3+ Treg cells, and promoted Th17 cells. Thus, Gal-9/Tim-3 signaling pathway activation may protect against renal IRI by inhibiting Th17 cell production and inducing Foxp3+ Treg cell expansion. Our study suggests that the Gal-9/Tim-3 signaling pathway may be targeted by immunotherapy in renal IRI.
Collapse
|
21
|
Ramirez-Gonzalez JB, Morales-BuenRostro LE, Garcia-Covarrubias L, Pacheco-Domínguez RL, Durazo-Arvizu R, Cuevas-Medina EN, Furuzawa-Carballeda J, Osorio-Juárez RA, Correa-Rotter R, Rincón-Pedrero R, Alberú-Gómez J, López Cervantes M. Assessment of the Relationship Between Inflammation and Glomerular Filtration Rate. Can J Kidney Health Dis 2023; 10:20543581221132748. [PMID: 36700057 PMCID: PMC9869199 DOI: 10.1177/20543581221132748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 08/22/2022] [Indexed: 01/20/2023] Open
Abstract
Background Chronic kidney disease (CKD) is a global health problem. As it progresses to end stages, renal replacement therapy is required but ultimately, the best treatment is transplantation. Decreased renal function has been associated with an inflammatory state associated to primary CKD and in kidney transplant recipients (KTRs). Objective To establish how the serum concentrations of some cytokines, such as interleukin (IL)-2, IL-8, IL-22, IL-17α, interferon-gamma, IL-4, and transforming growth factor-β, correlate with various CKD stages. Methods One hundred and forty-one KTRs between the ages of 18 and 75 years were included in the study. We also included 112 live kidney donors, 37 CKD PGCKD+3, and 76 GPhealthy. Participants were grouped according to their glomerular filtration rate (GFR) and their circulating cytokine levels, previously quantified by ELISA. Results By linear regression analysis, we established the relation of each cytokine with the GFR. Transforming growth factor-β correlated positively with the GFR in the study population, except in healthy individuals. A negative correlation of IL-8 and IL-17α and GFR was found in all cases. Conclusions Whether these cytokines (IL-8 and IL-17α) could be used as inflammatory biomarkers indicating CKD progression, regardless of the type of population, remains to be prospectively determined.
Collapse
Affiliation(s)
- J. B. Ramirez-Gonzalez
- Faculty of Medicine, Department of Public Health, Universidad Nacional Autónoma de México, Ciudad de Mexico, México
| | - L. E. Morales-BuenRostro
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de Mexico, México
| | - L. Garcia-Covarrubias
- Department of Transplantation, Hospital General de México Dr. Eduardo Liceaga, Ciudad de México, México
| | - R. L. Pacheco-Domínguez
- Faculty of Medicine, Department of Public Health, Universidad Nacional Autónoma de México, Ciudad de Mexico, México
| | - R. Durazo-Arvizu
- Department of Public Health Sciences, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - E. N. Cuevas-Medina
- Department of Cardiology, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - J. Furuzawa-Carballeda
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de Mexico, México
| | - R. A. Osorio-Juárez
- Faculty of Medicine, Department of Public Health, Universidad Nacional Autónoma de México, Ciudad de Mexico, México
| | - R. Correa-Rotter
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de Mexico, México
| | - R. Rincón-Pedrero
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de Mexico, México
| | - Josefina Alberú-Gómez
- Department of Transplantation, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de Mexico, México,Josefina Alberú-Gómez, Department of Transplantation, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga # 15, Colonia Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México CP 14080, México.
| | - Malaquías López Cervantes
- Faculty of Medicine, Department of Public Health, Universidad Nacional Autónoma de México, Ciudad de Mexico, México,Malaquías López Cervantes, Faculty of Medicine, Department of Public Health, Universidad Nacional Autónoma de México, Av. Universidad Nacional Autónoma de México #3000, Colonica Universidad Nacional Autonoma de México, CU, Delegación Coyoacán, Ciudad de México, CP 04510, México. ;
| |
Collapse
|
22
|
Esnault S, Jarjour NN. Development of Adaptive Immunity and Its Role in Lung Remodeling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1426:287-351. [PMID: 37464127 DOI: 10.1007/978-3-031-32259-4_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Asthma is characterized by airflow limitations resulting from bronchial closure, which can be either reversible or fixed due to changes in airway tissue composition and structure, also known as remodeling. Airway remodeling is defined as increased presence of mucins-producing epithelial cells, increased thickness of airway smooth muscle cells, angiogenesis, increased number and activation state of fibroblasts, and extracellular matrix (ECM) deposition. Airway inflammation is believed to be the main cause of the development of airway remodeling in asthma. In this chapter, we will review the development of the adaptive immune response and the impact of its mediators and cells on the elements defining airway remodeling in asthma.
Collapse
|
23
|
Estrogen Protects against Renal Ischemia-Reperfusion Injury by Regulating Th17/Treg Cell Immune Balance. DISEASE MARKERS 2022; 2022:7812099. [PMID: 36246554 PMCID: PMC9560860 DOI: 10.1155/2022/7812099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/01/2022] [Accepted: 09/23/2022] [Indexed: 12/31/2022]
Abstract
Inflammation is a critical mediator of renal ischemia-reperfusion (I/R) injury (IRI), and T lymphocytes exert a key role in the renal IRI-induced inflammation. Connexin 43 (Cx43) is related to the maintenance of T lymphocyte homeostasis. Various preclinical researches have reported that estrogen is a renoprotective agent based on its anti-inflammatory potential. The present research is aimed at studying the role of T lymphocytes activated by Cx43 in 17β-estradiol-mediated protection against renal IRI. Female rats were classified into six groups: control rats, I/R rats, ovariectomized rats, ovariectomized I/R rats, and ovariectomized rats treated with 17β-estradiol or gap27. Levels of serum creatinine (Scr) and blood urea nitrogen (BUN) and Paller scoring were dramatically increased in I/R rats, especially in ovariectomized rats. By contrast, these indicators were markedly decreased by administering estradiol or gap27. Immunofluorescence staining revealed that CD4+ T cells infiltrated kidney tissues in the early stage of IRI. In both peripheral blood and renal tissue, the proportion of CD3+CD4+ T cells and ratio of CD4+ to CD8+ were high in I/R rats, especially in ovariectomized rats. The proportion of CD3+CD8+ T cells was low in peripheral blood but high in renal tissues. Administration of estrogen or Gap27 reversed these effects. IL-17 levels in both serum and tissue homogenate were significantly increased in ovariectomized rats subjected to I/R but significantly decreased in estrogen or gap 27 treated rats. The opposite trend was observed for IL-10 levels. Correlation analysis demonstrated that IL-17 was correlated positively with BUN, Scr, and Paller scores, while IL-10 was negatively correlated with these indicators. Western blot showed that Cx43 expression was markedly increased in the peripheral blood T lymphocytes of I/R rats, especially ovariectomized rats. After intervention with estrogen and gap27, Cx43 expression was significantly downregulated. These findings indicate that Cx43 may participate in the regulation of Th17/Treg balance by estrogen against renal IRI.
Collapse
|
24
|
Yan P, Ke B, Fang X. Ion channels as a therapeutic target for renal fibrosis. Front Physiol 2022; 13:1019028. [PMID: 36277193 PMCID: PMC9581181 DOI: 10.3389/fphys.2022.1019028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Renal ion channel transport and electrolyte disturbances play an important role in the process of functional impairment and fibrosis in the kidney. It is well known that there are limited effective drugs for the treatment of renal fibrosis, and since a large number of ion channels are involved in the renal fibrosis process, understanding the mechanisms of ion channel transport and the complex network of signaling cascades between them is essential to identify potential therapeutic approaches to slow down renal fibrosis. This review summarizes the current work of ion channels in renal fibrosis. We pay close attention to the effect of cystic fibrosis transmembrane conductance regulator (CFTR), transmembrane Member 16A (TMEM16A) and other Cl− channel mediated signaling pathways and ion concentrations on fibrosis, as well as the various complex mechanisms for the action of Ca2+ handling channels including Ca2+-release-activated Ca2+ channel (CRAC), purinergic receptor, and transient receptor potential (TRP) channels. Furthermore, we also focus on the contribution of Na+ transport such as epithelial sodium channel (ENaC), Na+, K+-ATPase, Na+-H+ exchangers, and K+ channels like Ca2+-activated K+ channels, voltage-dependent K+ channel, ATP-sensitive K+ channels on renal fibrosis. Proposed potential therapeutic approaches through further dissection of these mechanisms may provide new therapeutic opportunities to reduce the burden of chronic kidney disease.
Collapse
|
25
|
Dhital R, Anand S, Graber B, Zeng Q, Velazquez VM, Boddeda SR, Fitch JR, Minz RW, Minz M, Sharma A, Cianciolo R, Shimamura M. Murine cytomegalovirus promotes renal allograft inflammation via Th1/17 cells and IL-17A. Am J Transplant 2022; 22:2306-2322. [PMID: 35671112 PMCID: PMC9547825 DOI: 10.1111/ajt.17116] [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: 11/09/2021] [Revised: 05/24/2022] [Accepted: 06/02/2022] [Indexed: 01/25/2023]
Abstract
Human cytomegalovirus (HCMV) infection is associated with renal allograft failure. Allograft damage in animal models is accelerated by CMV-induced T helper 17 (Th17) cell infiltrates. However, the mechanisms whereby CMV promotes Th17 cell-mediated pathological organ inflammation are uncharacterized. Here we demonstrate that murine CMV (MCMV)-induced intragraft Th17 cells have a Th1/17 phenotype co-expressing IFN-γ and/or TNF-α, but only a minority of these cells are MCMV specific. Instead, MCMV promotes intragraft expression of CCL20 and CXCL10, which are associated with recruitment of CCR6+ CXCR3+ Th17 cells. MCMV also enhances Th17 cell infiltrates after ischemia-reperfusion injury, independent of allogeneic responses. Pharmacologic inhibition of the Th17 cell signature cytokine, IL-17A, ameliorates MCMV-associated allograft damage without increasing intragraft viral loads or reducing MCMV-specific Th1 cell infiltrates. Clinically, HCMV DNAemia is associated with higher serum IL-17A among renal transplant patients with acute rejection, linking HCMV reactivation with Th17 cell cytokine expression. In summary, CMV promotes allograft damage via cytokine-mediated Th1/17 cell recruitment, which may be pharmacologically targeted to mitigate graft injury while preserving antiviral T cell immunity.
Collapse
Affiliation(s)
- Ravi Dhital
- Center for Vaccines and Immunity, The Abigail Wexner Research InstituteNationwide Children's HospitalColumbusOhioUSA
| | - Shashi Anand
- Department of ImmunopathologyPost Graduate Institute of Medical Education and Research (PGIMER)ChandigarhIndia
| | - Brianna Graber
- Center for Vaccines and Immunity, The Abigail Wexner Research InstituteNationwide Children's HospitalColumbusOhioUSA
| | - Qiang Zeng
- Center for Regenerative MedicineThe Abigail Research Institute, Nationwide Children's HospitalColumbusOhioUSA
| | - Victoria M. Velazquez
- Center for Vaccines and Immunity, The Abigail Wexner Research InstituteNationwide Children's HospitalColumbusOhioUSA
| | - Srinivasa R. Boddeda
- Center for Vaccines and Immunity, The Abigail Wexner Research InstituteNationwide Children's HospitalColumbusOhioUSA
| | - James R. Fitch
- The Steve and Cindy Rasmussen Institute for Genomic MedicineNationwide Children's HospitalColumbusOhioUSA
| | - Ranjana W. Minz
- Department of ImmunopathologyPost Graduate Institute of Medical Education and Research (PGIMER)ChandigarhIndia
| | - Mukut Minz
- Department of Renal Transplant SurgeryPost Graduate Institute of Medical Education and Research (PGIMER)ChandigarhIndia
| | - Ashish Sharma
- Department of Renal Transplant SurgeryPost Graduate Institute of Medical Education and Research (PGIMER)ChandigarhIndia
| | - Rachel Cianciolo
- Department of Veterinary Biosciences, College of Veterinary MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Masako Shimamura
- Center for Vaccines and Immunity, The Abigail Wexner Research InstituteNationwide Children's HospitalColumbusOhioUSA,Division of Pediatric Infectious Diseases, Department of Pediatrics, College of MedicineThe Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
26
|
Th17/Regulatory T-Cell Imbalance and Acute Kidney Injury in Patients with Sepsis. J Clin Med 2022; 11:jcm11144027. [PMID: 35887790 PMCID: PMC9320223 DOI: 10.3390/jcm11144027] [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: 06/01/2022] [Revised: 06/28/2022] [Accepted: 07/08/2022] [Indexed: 12/10/2022] Open
Abstract
To analyze the predictive value of the Th17/Treg ratio for renal injury in sepsis patients, a prospective observational study was conducted. Adult patients with sepsis were enrolled and divided into a sepsis-induced acute kidney injury (SAKI) group and a sepsis-without-AKI group. Logistic regression was used to analyze the independent predictors of SAKI, and the ROC curve was plotted to evaluate the predictive value of the Th17/Treg ratio for renal injury in patients with sepsis. A total of 124 patients were enrolled in this study, including 60 cases (48.39%) of SAKI. Patients who developed sepsis-induced acute kidney injury had a higher Th17/Treg ratio level compared to patients without it (0.11 [0.07, 0.28] versus 0.06 [0.05, 0.16], p < 0.05, respectively. The area under the receiver operating characteristic curve of the Th17/Treg ratio to predict sepsis-induced acute kidney injury was 0.669 (95% CI 0.574−0.763, p < 0.05). The Th17/Treg ratio was associated with SAKI (OR 1.15, 95%CI [1.06−1.24], p < 0.05, non-adjusted and R 1.12, 95%CI [1.00−1.25], p < 0.05, adjusted). The use of the Th17/Treg ratio improved the prediction performance of the prediction model of NAGL. The median Th17/Treg ratio significantly increased with the stratified KDIGO stage (p < 0.05). Th17/Treg imbalance was associated with occurrence of acute kidney injury and AKI severity in patients with sepsis. The Th17/Treg ratio could be a potential predictive marker of sepsis-induced acute kidney injury.
Collapse
|
27
|
Magkrioti C, Antonopoulou G, Fanidis D, Pliaka V, Sakellaropoulos T, Alexopoulos LG, Ullmer C, Aidinis V. Lysophosphatidic Acid Is a Proinflammatory Stimulus of Renal Tubular Epithelial Cells. Int J Mol Sci 2022; 23:ijms23137452. [PMID: 35806457 PMCID: PMC9267536 DOI: 10.3390/ijms23137452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/28/2022] [Accepted: 07/02/2022] [Indexed: 02/01/2023] Open
Abstract
Chronic kidney disease (CKD) refers to a spectrum of diseases defined by renal fibrosis, permanent alterations in kidney structure, and low glomerular-filtration rate. Prolonged epithelial-tubular damage involves a series of changes that eventually lead to CKD, highlighting the importance of tubular epithelial cells in this process. Lysophosphatidic acid (LPA) is a bioactive lipid that signals mainly through its six cognate LPA receptors and is implicated in several chronic inflammatory pathological conditions. In this report, we have stimulated human proximal tubular epithelial cells (HKC-8) with LPA and 175 other possibly pathological stimuli, and simultaneously detected the levels of 27 intracellular phosphoproteins and 32 extracellular secreted molecules with multiplex ELISA. This quantification revealed a large amount of information concerning the signaling and the physiology of HKC-8 cells that can be extrapolated to other proximal tubular epithelial cells. LPA responses clustered with pro-inflammatory stimuli such as TNF and IL-1, promoting the phosphorylation of important inflammatory signaling hubs, including CREB1, ERK1, JUN, IκΒα, and MEK1, as well as the secretion of inflammatory factors of clinical relevance, including CCL2, CCL3, CXCL10, ICAM1, IL-6, and IL-8, most of them shown for the first time in proximal tubular epithelial cells. The identified LPA-induced signal-transduction pathways, which were pharmacologically validated, and the secretion of the inflammatory factors offer novel insights into the possible role of LPA in CKD pathogenesis.
Collapse
Affiliation(s)
- Christiana Magkrioti
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece; (C.M.); (G.A.); (D.F.)
| | - Georgia Antonopoulou
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece; (C.M.); (G.A.); (D.F.)
| | - Dionysios Fanidis
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece; (C.M.); (G.A.); (D.F.)
| | - Vaia Pliaka
- ProtATonce Ltd., 15343 Athens, Greece; (V.P.); (T.S.); (L.G.A.)
| | | | - Leonidas G. Alexopoulos
- ProtATonce Ltd., 15343 Athens, Greece; (V.P.); (T.S.); (L.G.A.)
- School of Mechanical Engineering, National Technical University of Athens, 15780 Zografou, Greece
| | - Christoph Ullmer
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland;
| | - Vassilis Aidinis
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece; (C.M.); (G.A.); (D.F.)
- Correspondence:
| |
Collapse
|
28
|
Fernandez-Ruiz R, Belmont HM. The role of anticomplement therapy in lupus nephritis. Transl Res 2022; 245:1-17. [PMID: 35158097 DOI: 10.1016/j.trsl.2022.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/03/2022] [Accepted: 02/07/2022] [Indexed: 10/19/2022]
Abstract
The complement system plays crucial roles in homeostasis and host defense against microbes. Deficiency of early complement cascade components has been associated with increased susceptibility to systemic lupus erythematosus (SLE), whereas excessive complement consumption is a hallmark of this disease. Although enhanced classical pathway activation by immune complexes was initially thought to be the main contributor to lupus nephritis (LN) pathogenesis, an increasing body of evidence has suggested the alternative and the lectin pathways are also involved. Therapeutic agents targeting complement activation have been used in LN patients and clinical trials are ongoing. We review the mechanisms by which complement system dysregulation contributes to renal injury in SLE and summarize the latest evidence on the use of anticomplement agents to manage this condition.
Collapse
Affiliation(s)
- Ruth Fernandez-Ruiz
- Division of Rheumatology, NYU Grossman School of Medicine, New York, New York
| | | |
Collapse
|
29
|
Lee K, Jang HR. Role of T cells in ischemic acute kidney injury and repair. Korean J Intern Med 2022; 37:534-550. [PMID: 35508946 PMCID: PMC9082442 DOI: 10.3904/kjim.2021.526] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 01/11/2022] [Indexed: 11/27/2022] Open
Abstract
Ischemic acute kidney injury (AKI) is a common medical problem with significant mortality and morbidity, affecting a large number of patients globally. Ischemic AKI is associated with intrarenal inflammation as well as systemic inflammation; thus, the innate and adaptive immune systems are implicated in the pathogenesis of ischemic AKI. Among various intrarenal immune cells, T cells play major roles in the injury process and in the repair mechanism affecting AKI to chronic kidney disease transition. Importantly, T cells also participate in distant organ crosstalk during AKI, which affects the overall outcomes. Therefore, targeting T cell-mediated pathways and T cell-based therapies have therapeutic promise for ischemic AKI. Here, we review the major populations of kidney T cells and their roles in ischemic AKI.
Collapse
Affiliation(s)
- Kyungho Lee
- Nephrology Division, Department of Medicine, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hye Ryoun Jang
- Nephrology Division, Department of Medicine, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
30
|
Collett JA, Ortiz-Soriano V, Li X, Flannery AH, Toto RD, Moe OW, Basile DP, Neyra JA. Serum IL-17 levels are higher in critically ill patients with AKI and associated with worse outcomes. Crit Care 2022; 26:107. [PMID: 35422004 PMCID: PMC9008961 DOI: 10.1186/s13054-022-03976-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 04/03/2022] [Indexed: 11/30/2022] Open
Abstract
Background Interleukin-17 (IL-17) antagonism in rats reduces the severity and progression of AKI. IL-17-producing circulating T helper-17 (TH17) cells is increased in critically ill patients with AKI indicating that this pathway is also activated in humans. We aim to compare serum IL-17A levels in critically ill patients with versus without AKI and to examine their relationship with mortality and major adverse kidney events (MAKE). Methods Multicenter, prospective study of ICU patients with AKI stage 2 or 3 and without AKI. Samples were collected at 24–48 h after AKI diagnosis or ICU admission (in those without AKI) [timepoint 1, T1] and 5–7 days later [timepoint 2, T2]. MAKE was defined as the composite of death, dependence on kidney replacement therapy or a reduction in eGFR of ≥ 30% from baseline up to 90 days following hospital discharge. Results A total of 299 patients were evaluated. Patients in the highest IL-17A tertile (versus lower tertiles) at T1 had higher acuity of illness and comorbidity scores. Patients with AKI had higher levels of IL-17A than those without AKI: T1 1918.6 fg/ml (692.0–5860.9) versus 623.1 fg/ml (331.7–1503.4), p < 0.001; T2 2167.7 fg/ml (839.9–4618.9) versus 1193.5 fg/ml (523.8–2198.7), p = 0.006. Every onefold higher serum IL-17A at T1 was independently associated with increased risk of hospital mortality (aOR 1.35, 95% CI: 1.06–1.73) and MAKE (aOR 1.26, 95% CI: 1.02–1.55). The highest tertile of IL-17A (vs. the lowest tertile) was also independently associated with higher risk of MAKE (aOR 3.03, 95% CI: 1.34–6.87). There was no effect modification of these associations by AKI status. IL-17A levels remained significantly elevated at T2 in patients that died or developed MAKE. Conclusions Serum IL-17A levels measured by the time of AKI diagnosis or ICU admission were differentially elevated in critically ill patients with AKI when compared to those without AKI and were independently associated with hospital mortality and MAKE. Supplementary Information The online version contains supplementary material available at 10.1186/s13054-022-03976-4.
Collapse
|
31
|
Thomas BB, Lin B, Martinez-Camarillo JC, Zhu D, McLelland BT, Nistor G, Keirstead HS, Humayun MS, Seiler MJ. Co-grafts of Human Embryonic Stem Cell Derived Retina Organoids and Retinal Pigment Epithelium for Retinal Reconstruction in Immunodeficient Retinal Degenerate Royal College of Surgeons Rats. Front Neurosci 2021; 15:752958. [PMID: 34764853 PMCID: PMC8576198 DOI: 10.3389/fnins.2021.752958] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/04/2021] [Indexed: 12/15/2022] Open
Abstract
End-stage age-related macular degeneration (AMD) and retinitis pigmentosa (RP) are two major retinal degenerative (RD) conditions that result in irreversible vision loss. Permanent eye damage can also occur in battlefields or due to accidents. This suggests there is an unmet need for developing effective strategies for treating permanent retinal damages. In previous studies, co-grafted sheets of fetal retina with its retinal pigment epithelium (RPE) have demonstrated vision improvement in rat retinal disease models and in patients, but this has not yet been attempted with stem-cell derived tissue. Here we demonstrate a cellular therapy for irreversible retinal eye injuries using a "total retina patch" consisting of retinal photoreceptor progenitor sheets and healthy RPE cells on an artificial Bruch's membrane (BM). For this, retina organoids (ROs) (cultured in suspension) and polarized RPE sheets (cultured on an ultrathin parylene substrate) were made into a co-graft using bio-adhesives [gelatin, growth factor-reduced matrigel, and medium viscosity (MVG) alginate]. In vivo transplantation experiments were conducted in immunodeficient Royal College of Surgeons (RCS) rats at advanced stages of retinal degeneration. Structural reconstruction of the severely damaged retina was observed based on histological assessments and optical coherence tomography (OCT) imaging. Visual functional assessments were conducted by optokinetic behavioral testing and superior colliculus electrophysiology. Long-term survival of the co-graft in the rat subretinal space and improvement in visual function were observed. Immunohistochemistry showed that co-grafts grew, generated new photoreceptors and developed neuronal processes that were integrated into the host retina. This novel approach can be considered as a new therapy for complete replacement of a degenerated retina.
Collapse
Affiliation(s)
- Biju B. Thomas
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, United States
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA, United States
| | - Bin Lin
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, Irvine, CA, United States
- Stem Cell Research Center, University of California, Irvine, Irvine, CA, United States
| | - Juan Carlos Martinez-Camarillo
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, United States
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA, United States
| | - Danhong Zhu
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, United States
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Bryce T. McLelland
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, Irvine, CA, United States
- Stem Cell Research Center, University of California, Irvine, Irvine, CA, United States
| | | | | | - Mark S. Humayun
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, United States
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA, United States
| | - Magdalene J. Seiler
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, Irvine, CA, United States
- Stem Cell Research Center, University of California, Irvine, Irvine, CA, United States
- Department of Ophthalmology, University of California, Irvine, Irvine, CA, United States
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
32
|
Muniz IPR, Galantini MPL, Ribeiro IS, Gonçalves CV, Dos Santos DP, Moura TC, Silva ES, Silva NR, Cipriano BP, Correia TML, de Jesus Soares T, de Freitas LM, Costa DJ, da Silva RAA. Antimicrobial photodynamic therapy (aPDT) with curcumin controls intradermal infection by Staphylococcus aureus in mice with type 1 diabetes mellitus: a pilot study. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2021; 224:112325. [PMID: 34598018 DOI: 10.1016/j.jphotobiol.2021.112325] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is one of the main pathogens that cause infections in diabetic individuals. In this paper, we report the outcomes of our investigation on the intradermal application of antimicrobial photodynamic therapy (PDT) with curcumin in an infection induced by MRSA ATCC 43300 strain in the ear of mice with Type 1 Diabetes Mellitus (T1DM). A solution containing 100 μg of curcumin was photoactivated ex vivo with a LED light (450 nm) delivering a fluency of 13.5 J/cm3. This solution was administered in the ear intradermally, at the same inoculum site as the MRSA ATCC 43300 strain (PDT Group). This study also included the use of two control groups (both infected): One was treated with saline and the other was treated with non-photoactivated curcumin. The animals were euthanized 24 h after these treatments and samples of draining lymph node and treated ear were collected for examination. The PDT group showed lower bacterial load in the draining lymph node when compared to the saline and curcumin groups (p-value <0.05) 24 h after treatment. In addition to bacterial load, the PDT group presented a higher concentration of nitrates and nitrites in the draining lymph node when compared to the saline and curcumin groups (p-value <0.001). Examining the infectious site, despite apparently having similar inflammatory cell recruitment compared with the control groups, the PDT group showed a profile with less intense activity in the myeloperoxidase expression when compared to the saline group (p-value <0.001). Additionally, the detected concentration of cytokines such as IL-1β, IL-12, and IL-10 was significantly lower in the PDT group when compared to the saline group (p-value <0.01; p-value <0.05; p-value <0.05, respectively), thus presenting a less intense inflammatory response during infection resolution. Our pilot study showed for the first time the therapeutic potential of PDT using curcumin when administered intradermally in the treatment of infections caused by S. aureus in mice with T1DM.
Collapse
Affiliation(s)
- Igor Pereira Ribeiro Muniz
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Maria Poliana Leite Galantini
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Israel Souza Ribeiro
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil; Universidade Federal do Sul da Bahia, Campus Paulo Freire, 250 Praça Joana Angélica, Bairro São José, 45.988-058, Teixeira de Freitas, Bahia, Brazil
| | - Caroline Vieira Gonçalves
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Denisar Palmito Dos Santos
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Tatyana Chagas Moura
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Emely Soares Silva
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Nathalia Rosa Silva
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Barbara Porto Cipriano
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Thiago Macêdo Lopes Correia
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Telma de Jesus Soares
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Leandro Martins de Freitas
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil
| | - Dirceu Joaquim Costa
- Universidade Estadual do Sudoeste da Bahia, Campus Vitória da Conquista, Av. Edmundo Silveira Flores, 27-43 - Lot, Alto da Boa Vista, CEP: 45029-066 Vitória da Conquista, Bahia, Brasil
| | - Robson Amaro Augusto da Silva
- Universidade Federal da Bahia, Campus Anísio Teixeira - Instituto Multidisciplinar em Saúde, Rua Rio de Contas, 58, Bairro Candeias, CEP: 45.029-094 Vitória da Conquista, Bahia, Brasil.
| |
Collapse
|
33
|
Cao C, Yao Y, Zeng R. Lymphocytes: Versatile Participants in Acute Kidney Injury and Progression to Chronic Kidney Disease. Front Physiol 2021; 12:729084. [PMID: 34616308 PMCID: PMC8488268 DOI: 10.3389/fphys.2021.729084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/19/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Acute kidney injury (AKI) remains a major global public health concern due to its high morbidity and mortality. The progression from AKI to chronic kidney disease (CKD) makes it a scientific problem to be solved. However, it is with lack of effective treatments. Summary: Both innate and adaptive immune systems participate in the inflammatory process during AKI, and excessive or dysregulated immune responses play a pathogenic role in renal fibrosis, which is an important hallmark of CKD. Studies on the pathogenesis of AKI and CKD have clarified that renal injury induces the production of various chemokines by renal parenchyma cells or resident immune cells, which recruits multiple-subtype lymphocytes in circulation. Some infiltrated lymphocytes exacerbate injury by proinflammatory cytokine production, cytotoxicity, and interaction with renal resident cells, which constructs the inflammatory environment and induces further injury, even death of renal parenchyma cells. Others promote tissue repair by producing protective cytokines. In this review, we outline the diversity of these lymphocytes and their mechanisms to regulate the whole pathogenic stages of AKI and CKD; discuss the chronological responses and the plasticity of lymphocytes related to AKI and CKD progression; and introduce the potential therapies targeting lymphocytes of AKI and CKD, including the interventions of chemokines, cytokines, and lymphocyte frequency regulation in vivo, adaptive transfer of ex-expanded lymphocytes, and the treatments of gut microbiota or metabolite regulations based on gut-kidney axis. Key Message: In the process of AKI and CKD, T helper (Th) cells, innate, and innate-like lymphocytes exert mainly pathogenic roles, while double-negative T (DNT) cells and regulatory T cells (Tregs) are confirmed to be protective. Understanding the mechanisms by which lymphocytes mediate renal injury and renal fibrosis is necessary to promote the development of specific therapeutic strategies to protect from AKI and prevent the progression of CKD.
Collapse
Affiliation(s)
| | - Ying Yao
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Zeng
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
34
|
Memi G, Yazgan B. Adropin and spexin hormones regulate the systemic inflammation in adenine-induced chronic kidney failure in rat. CHINESE J PHYSIOL 2021; 64:194-201. [PMID: 34472450 DOI: 10.4103/cjp.cjp_13_21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Chronic kidney disease is one of the major global health problems. Chronic renal failure is stimulated by many cytokines and chemokines. Adropin and spexin (SPX) are peptides hormones. These peptides could affect inflammatory conditions, but this is unclear. Due to the limited information, we planned to investigate the impact of adropin and SPX hormones on systemic inflammation in adenine induced chronic kidney failure rat model. Chronic kidney failure was induced by administering adenine hemisulfate. Renal functions were measured by an autoanalyzer. Granulocyte colony-stimulating factor (G-CSF), interferon-gamma (IFN-γ), interleukin (IL)-1β, IL-2, IL-4, IL-5, IL-10, IL-12, IL-13, IL-17A, tumor necrosis factor-alpha, Eotaxin, growth-regulated oncogene-alpha, IP-10, monocyte chemoattractant protein (MCP)-1, MCP-3, macrophage inflammatory protein (MIP)-1α, MIP-2, and RANTES levels were determined by Luminex. We observed an increase in 24-h urine volume and serum creatinine. Blood urea nitrogen (BUN) and urine protein levels were also significantly higher in the chronic kidney failure (CKF) group. Urine protein and 24-h urine volume were reduced with adropin and SPX treatments. Furthermore, G-CSF, IFN-γ, IL-4, IL-5, IL-10, IL-12, IL-17A, and GRO-α significantly increased by CKF induction; however, these cytokines and chemokines significantly decreased by adropin treatment in the CKF group. Furthermore, adropin increased IP-10, MCP-1, MIP-1α, and MIP-2 levels. In addition, SPX treatment had a more limited effect, decreasing only G-CSF, IFN-γ, and IL-5 levels. The combined adropin + SPX treatment significantly reduced G-CSF, IFN-γ, IL-4, IL-5, IL-12, and IL-17A. Furthermore, IP-10, MCP-1, MCP-3, and MIP-2 were significantly increased by these combined treatments. Our findings indicate that renal functions and inflammatory response were modulated by adropin and SPX peptides. These peptides may have protective effects on systemic inflammation and renal failure progression.
Collapse
Affiliation(s)
- Gulsun Memi
- Department of Physiology, School of Medicine, Adıyaman University, Adıyaman, Turkey
| | - Burak Yazgan
- Department of Medical Services and Techniques, Sabuncuoglu Serefeddin Health Services Vocational School; Department of Molecular Medicine, Institute of Health Sciences, Amasya University, Amasya, Turkey
| |
Collapse
|
35
|
Paquissi FC, Abensur H. The Th17/IL-17 Axis and Kidney Diseases, With Focus on Lupus Nephritis. Front Med (Lausanne) 2021; 8:654912. [PMID: 34540858 PMCID: PMC8446428 DOI: 10.3389/fmed.2021.654912] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 08/04/2021] [Indexed: 12/28/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a disease characterized by dysregulation and hyperreactivity of the immune response at various levels, including hyperactivation of effector cell subtypes, autoantibodies production, immune complex formation, and deposition in tissues. The consequences of hyperreactivity to the self are systemic and local inflammation and tissue damage in multiple organs. Lupus nephritis (LN) is one of the most worrying manifestations of SLE, and most patients have this involvement at some point in the course of the disease. Among the effector cells involved, the Th17, a subtype of T helper cells (CD4+), has shown significant hyperactivation and participates in kidney damage and many other organs. Th17 cells have IL-17A and IL-17F as main cytokines with receptors expressed in most renal cells, being involved in the activation of many proinflammatory and profibrotic pathways. The Th17/IL-17 axis promotes and maintains repetitive tissue damage and maladaptive repair; leading to fibrosis, loss of organ architecture and function. In the podocytes, the Th17/IL-17 axis effects include changes of the cytoskeleton with increased motility, decreased expression of health proteins, increased oxidative stress, and activation of the inflammasome and caspases resulting in podocytes apoptosis. In renal tubular epithelial cells, the Th17/IL-17 axis promotes the activation of profibrotic pathways such as increased TGF-β expression and epithelial-mesenchymal transition (EMT) with consequent increase of extracellular matrix proteins. In addition, the IL-17 promotes a proinflammatory environment by stimulating the synthesis of inflammatory cytokines by intrinsic renal cells and immune cells, and the synthesis of growth factors and chemokines, which together result in granulopoiesis/myelopoiesis, and further recruitment of immune cells to the kidney. The purpose of this work is to present the prognostic and immunopathologic role of the Th17/IL-17 axis in Kidney diseases, with a special focus on LN, including its exploration as a potential immunotherapeutic target in this complication.
Collapse
Affiliation(s)
- Feliciano Chanana Paquissi
- Department of Medicine, Clínica Girassol, Luanda, Angola
- Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Hugo Abensur
- Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
36
|
Lee SH, Kim M, Han KD, Lee JH. Low hemoglobin levels and an increased risk of psoriasis in patients with chronic kidney disease. Sci Rep 2021; 11:14741. [PMID: 34285267 PMCID: PMC8292392 DOI: 10.1038/s41598-021-94165-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 07/07/2021] [Indexed: 02/08/2023] Open
Abstract
Chronic diseases, such as chronic kidney disease (CKD), are frequently accompanied by various comorbidities, including anemia, which is considered a surrogate marker of systemic inflammation. Psoriasis is a chronic inflammatory skin disease prevalent in patients with chronic disease. Psoriasis risk in patients with CKD, however, especially in patients with low hemoglobin levels, has never been investigated. In this study, we investigated associations between low hemoglobin levels and psoriasis in patients with CKD using data from the National Health Insurance Service of Korea. During a mean follow-up period of 6.16 ± 1.02 years, psoriasis was recorded in 13,803 patients with CKD (2.39% of CKD patients). The cumulative incidence of psoriasis was significantly higher in CKD patients with anemia (hemoglobin levels < 13 g/dL in men and < 12 g/dL in women) than those without. In multivariate-adjusted Cox proportional hazards regression models, the risk of psoriasis was significantly higher in anemic CKD patients than nonanemic CKD patients (hazard ratio [HR] 1.136, 95% CI 1.089–1.185, p < 0.001). Additionally, we noted that the incidence of psoriasis decreased with increasing hemoglobin levels in CKD patients (HR 0.953, 95% CI 0.942–0.965, p < 0.001). Altogether, our findings indicate that low hemoglobin levels are significantly related to psoriasis risk in patients with CKD. Further study is required to elucidate whether low hemoglobin levels have an impact on the development of psoriasis or are merely a surrogate marker of psoriasis risk in patients with CKD.
Collapse
Affiliation(s)
- Si-Hyung Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Korea
| | - Miri Kim
- Department of Dermatology, College of Medicine, Yeouido St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Kyung-Do Han
- Department of Medical Statistics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ji Hyun Lee
- Department of Dermatology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Korea.
| |
Collapse
|
37
|
Cantero-Navarro E, Fernández-Fernández B, Ramos AM, Rayego-Mateos S, Rodrigues-Diez RR, Sánchez-Niño MD, Sanz AB, Ruiz-Ortega M, Ortiz A. Renin-angiotensin system and inflammation update. Mol Cell Endocrinol 2021; 529:111254. [PMID: 33798633 DOI: 10.1016/j.mce.2021.111254] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 02/05/2021] [Accepted: 03/18/2021] [Indexed: 12/12/2022]
Abstract
The most classical view of the renin-angiotensin system (RAS) emphasizes its role as an endocrine regulator of sodium balance and blood pressure. However, it has long become clear that the RAS has pleiotropic actions that contribute to organ damage, including modulation of inflammation. Angiotensin II (Ang II) activates angiotensin type 1 receptors (AT1R) to promote an inflammatory response and organ damage. This represents the pathophysiological basis for the successful use of RAS blockers to prevent and treat kidney and heart disease. However, other RAS components could have a built-in capacity to brake proinflammatory responses. Angiotensin type 2 receptor (AT2R) activation can oppose AT1R actions, such as vasodilatation, but its involvement in modulation of inflammation has not been conclusively proven. Angiotensin-converting enzyme 2 (ACE2) can process Ang II to generate angiotensin-(1-7) (Ang-(1-7)), that activates the Mas receptor to exert predominantly anti-inflammatory responses depending on the context. We now review recent advances in the understanding of the interaction of the RAS with inflammation. Specific topics in which novel information became available recently include intracellular angiotensin receptors; AT1R posttranslational modifications by tissue transglutaminase (TG2) and anti-AT1R autoimmunity; RAS modulation of lymphoid vessels and T lymphocyte responses, especially of Th17 and Treg responses; interactions with toll-like receptors (TLRs), programmed necrosis, and regulation of epigenetic modulators (e.g. microRNAs and bromodomain and extraterminal domain (BET) proteins). We additionally discuss an often overlooked effect of the RAS on inflammation which is the downregulation of anti-inflammatory factors such as klotho, peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α), transient receptor potential ankyrin 1 (TRPA1), SNF-related serine/threonine-protein kinase (SNRK), serine/threonine-protein phosphatase 6 catalytic subunit (Ppp6C) and n-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP). Both transcription factors, such as nuclear factor κB (NF-κB), and epigenetic regulators, such as miRNAs are involved in downmodulation of anti-inflammatory responses. A detailed analysis of pathways and targets for downmodulation of anti-inflammatory responses constitutes a novel frontier in RAS research.
Collapse
Affiliation(s)
- Elena Cantero-Navarro
- Molecular and Cellular Biology in Renal and Vascular Pathology. IIS-Fundación Jiménez Díaz-Universidad Autónoma, Madrid, Spain; Red de Investigación Renal (REDINREN), Spain
| | - Beatriz Fernández-Fernández
- Red de Investigación Renal (REDINREN), Spain; Unidad de Diálisis. IIS-Fundación Jiménez Díaz-Universidad Autónoma, Madrid, Spain
| | - Adrian M Ramos
- Red de Investigación Renal (REDINREN), Spain; Unidad de Diálisis. IIS-Fundación Jiménez Díaz-Universidad Autónoma, Madrid, Spain
| | - Sandra Rayego-Mateos
- Molecular and Cellular Biology in Renal and Vascular Pathology. IIS-Fundación Jiménez Díaz-Universidad Autónoma, Madrid, Spain; Red de Investigación Renal (REDINREN), Spain
| | - Raúl R Rodrigues-Diez
- Molecular and Cellular Biology in Renal and Vascular Pathology. IIS-Fundación Jiménez Díaz-Universidad Autónoma, Madrid, Spain; Red de Investigación Renal (REDINREN), Spain
| | - María Dolores Sánchez-Niño
- Red de Investigación Renal (REDINREN), Spain; Unidad de Diálisis. IIS-Fundación Jiménez Díaz-Universidad Autónoma, Madrid, Spain
| | - Ana B Sanz
- Red de Investigación Renal (REDINREN), Spain; Unidad de Diálisis. IIS-Fundación Jiménez Díaz-Universidad Autónoma, Madrid, Spain
| | - Marta Ruiz-Ortega
- Molecular and Cellular Biology in Renal and Vascular Pathology. IIS-Fundación Jiménez Díaz-Universidad Autónoma, Madrid, Spain; Red de Investigación Renal (REDINREN), Spain.
| | - Alberto Ortiz
- Red de Investigación Renal (REDINREN), Spain; Unidad de Diálisis. IIS-Fundación Jiménez Díaz-Universidad Autónoma, Madrid, Spain.
| |
Collapse
|
38
|
Basile DP, Ullah MM, Collet JA, Mehrotra P. T helper 17 cells in the pathophysiology of acute and chronic kidney disease. Kidney Res Clin Pract 2021; 40:12-28. [PMID: 33789382 PMCID: PMC8041630 DOI: 10.23876/j.krcp.20.185] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
Both acute and chronic kidney disease have a strong underlying inflammatory component. This review focuses primarily on T helper 17 (Th17) cells as mediators of inflammation and their potential to modulate acute and chronic kidney disease. We provide updated information on factors and signaling pathways that promote Th17 cell differentiation with specific reference to kidney disease. We highlight numerous clinical studies that have investigated Th17 cells in the setting of human kidney disease and provide updated summaries from various experimental animal models of kidney disease indicating an important role for Th17 cells in renal fibrosis and hypertension. We focus on the pleiotropic effects of Th17 cells in different renal cell types as potentially relevant to the pathogenesis of kidney disease. Finally, we highlight studies that present contrasting roles for Th17 cells in kidney disease progression.
Collapse
Affiliation(s)
- David P Basile
- Department of Anatomy, Cell Biology & Physiology, Indiana University of Medicine, Indianapolis, IN, United States
| | - Md Mahbub Ullah
- Department of Anatomy, Cell Biology & Physiology, Indiana University of Medicine, Indianapolis, IN, United States
| | - Jason A Collet
- Department of Anatomy, Cell Biology & Physiology, Indiana University of Medicine, Indianapolis, IN, United States
| | - Purvi Mehrotra
- Department of Anatomy, Cell Biology & Physiology, Indiana University of Medicine, Indianapolis, IN, United States
| |
Collapse
|
39
|
Basile DP, Abais-Battad JM, Mattson DL. Contribution of Th17 cells to tissue injury in hypertension. Curr Opin Nephrol Hypertens 2021; 30:151-158. [PMID: 33394732 PMCID: PMC8221512 DOI: 10.1097/mnh.0000000000000680] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Hypertension has been demonstrated to be a chief contributor to morbidity and mortality throughout the world. Although the cause of hypertension is multifactorial, emerging evidence, obtained in experimental studies, as well as observational studies in humans, points to the role of inflammation and immunity. Many aspects of immune function have now been implicated in hypertension and end-organ injury; this review will focus upon the recently-described role of Th17 cells in this pathophysiological response. RECENT FINDINGS Studies in animal models and human genetic studies point to a role in the adaptive immune system as playing a contributory role in hypertension and renal tissue damage. Th17 cells, which produce the cytokine IL17, are strongly pro-inflammatory cells, which may contribute to tissue damage if expressed in chronic disease conditions. The activity of these cells may be enhanced by physiological factors associated with hypertension such as dietary salt or Ang II. This activity may culminate in the increased sodium retaining activity and exacerbation of inflammation and renal fibrosis via multiple cellular mechanisms. SUMMARY Th17 cells are a distinct component of the adaptive immune system that may strongly enhance pathways leading to increased sodium reabsorption, elevated vascular tone and end-organ damage. Moreover, this pathway may lend itself towards specific targeting for treatment of kidney disease and hypertension.
Collapse
Affiliation(s)
- David P Basile
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - David L Mattson
- Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA
| |
Collapse
|
40
|
Tsugawa-Shimizu Y, Fujishima Y, Kita S, Minami S, Sakaue TA, Nakamura Y, Okita T, Kawachi Y, Fukada S, Namba-Hamano T, Takabatake Y, Isaka Y, Nishizawa H, Ranscht B, Maeda N, Shimomura I. Increased vascular permeability and severe renal tubular damage after ischemia-reperfusion injury in mice lacking adiponectin or T-cadherin. Am J Physiol Endocrinol Metab 2021; 320:E179-E190. [PMID: 33284092 PMCID: PMC8260375 DOI: 10.1152/ajpendo.00393.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Adiponectin (APN) is a circulating protein specifically produced by adipocytes. Native APN specifically binds to T-cadherin, a glycosylphosphatidylinositol-anchored protein, mediating the exosome-stimulating effects of APN in endothelial, muscle, and mesenchymal stem cells. It was previously reported that APN has beneficial effects on kidney diseases, but the role of T-cadherin has not been clarified yet. Here, our immunofluorescence study indicated the existence of both T-cadherin and APN protein in pericytes, subsets of tissue-resident mesenchymal stem/progenitor cells positive for platelet-derived growth factor receptor β (PDGFRβ), surrounding peritubular capillaries. In an acute renal ischemia-reperfusion (I/R) model, T-cadherin-knockout (Tcad-KO) mice, similar to APN-KO mice, exhibited the more progressive phenotype of renal tubular damage and increased vascular permeability than wild-type mice. In addition, in response to I/R-injury, the renal PDGFRβ-positive cell area increased in wild-type mice, but opposingly decreased in both Tcad-KO and APN-KO mice, suggesting severe pericyte loss. Mouse primary pericytes also expressed T-cadherin. APN promoted exosome secretion in a T-cadherin-dependent manner. Such exosome production from pericytes may play an important role in maintaining the capillary network and APN-mediated inhibition of renal tubular injury. In summary, our study suggested that APN protected the kidney in an acute renal injury model by binding to T-cadherin.NEW & NOTEWORTHY In the kidney, T-cadherin-associated adiponectin protein existed on peritubular capillary pericytes. In an acute renal ischemia-reperfusion model, deficiency of adiponectin or T-cadherin exhibited the more progressive phenotype of renal tubular damage and increased vascular permeability, accompanied by severe pericyte loss. In vitro, adiponectin promoted exosome secretion from mouse primary pericytes in a T-cadherin-dependent manner. Adiponectin plays an important role in maintaining the capillary network and amelioration of renal tubular injury by binding to T-cadherin.
Collapse
Affiliation(s)
- Yuri Tsugawa-Shimizu
- Department of Metabolic Medicine, Graduate School of Medicine and Faculty of Medicine, Osaka University, Suita, Japan
| | - Yuya Fujishima
- Department of Metabolic Medicine, Graduate School of Medicine and Faculty of Medicine, Osaka University, Suita, Japan
| | - Shunbun Kita
- Department of Metabolic Medicine, Graduate School of Medicine and Faculty of Medicine, Osaka University, Suita, Japan
- Department of Adipose Management, Graduate School of Medicine and Faculty of Medicine, Osaka University, Suita, Japan
| | - Satoshi Minami
- Department of Nephrology, Graduate School of Medicine and Faculty of Medicine, Osaka University, Suita, Japan
| | - Taka-Aki Sakaue
- Department of Metabolic Medicine, Graduate School of Medicine and Faculty of Medicine, Osaka University, Suita, Japan
| | - Yuto Nakamura
- Department of Metabolic Medicine, Graduate School of Medicine and Faculty of Medicine, Osaka University, Suita, Japan
| | - Tomonori Okita
- Department of Metabolic Medicine, Graduate School of Medicine and Faculty of Medicine, Osaka University, Suita, Japan
| | - Yusuke Kawachi
- Department of Metabolic Medicine, Graduate School of Medicine and Faculty of Medicine, Osaka University, Suita, Japan
| | - Shiro Fukada
- Department of Metabolic Medicine, Graduate School of Medicine and Faculty of Medicine, Osaka University, Suita, Japan
| | - Tomoko Namba-Hamano
- Department of Nephrology, Graduate School of Medicine and Faculty of Medicine, Osaka University, Suita, Japan
| | - Yoshitsugu Takabatake
- Department of Nephrology, Graduate School of Medicine and Faculty of Medicine, Osaka University, Suita, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Graduate School of Medicine and Faculty of Medicine, Osaka University, Suita, Japan
| | - Hitoshi Nishizawa
- Department of Metabolic Medicine, Graduate School of Medicine and Faculty of Medicine, Osaka University, Suita, Japan
| | - Barbara Ranscht
- Development, Aging and Regeneration Program, NIH-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Norikazu Maeda
- Department of Metabolic Medicine, Graduate School of Medicine and Faculty of Medicine, Osaka University, Suita, Japan
- Department of Metabolism and Atherosclerosis, Graduate School of Medicine and Faculty of Medicine, Osaka University, Suita, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine and Faculty of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
41
|
Lin X, Li J, Tan R, Zhong X, Yang J, Wang L. Identification of Hub Genes Associated with the Development of Acute Kidney Injury by Weighted Gene Co-Expression Network Analysis. Kidney Blood Press Res 2021; 46:63-73. [PMID: 33401265 DOI: 10.1159/000511661] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/17/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a severe clinical syndrome, causing a profound medical and socioeconomic burden worldwide. This study aimed to explore underlying molecular targets related to the progression of AKI. METHODS A public database originated from the NCBI GEO database (serial number: GSE121190) and a well-established and unbiased method of weighted gene co-expression network analysis (WGCNA) to identify hub genes and potential pathways were used. Furthermore, the unbiased hub genes were validated in 2 classic models of AKI in a rodent model: chemically established AKI by cisplatin- and ischemia reperfusion-induced AKI. RESULTS A total of 17 modules were finally obtained by the unbiased method of WGCNA, where the genes in turquoise module displayed strong correlation with the development of AKI. In addition, the results of gene ontology revealed that the genes in turquoise module were involved in renal injury and renal fibrosis. Thus, the hub genes were further validated by experimental methods and primarily obtained Rplp1 and Lgals1 as key candidate genes related to the progression of AKI by the advantage of quantitative PCR, Western blotting, and in situ tissue fluorescence. Importantly, the expression of Rplp1 and Lgals1 at the protein level showed positive correlation with renal function, including serum Cr and BUN. CONCLUSIONS By the advantage of unbiased bioinformatic method and consequent experimental verification, this study lays the foundation basis for the pathogenesis and therapeutic agent development of AKI.
Collapse
Affiliation(s)
- Xiao Lin
- Laboratory of Organ Fibrosis Prophylaxis and Treatment by Combine Traditional Chinese and Western Medicine, Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jianchun Li
- Laboratory of Organ Fibrosis Prophylaxis and Treatment by Combine Traditional Chinese and Western Medicine, Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Ruizhi Tan
- Laboratory of Organ Fibrosis Prophylaxis and Treatment by Combine Traditional Chinese and Western Medicine, Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xia Zhong
- Laboratory of Organ Fibrosis Prophylaxis and Treatment by Combine Traditional Chinese and Western Medicine, Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jieke Yang
- Laboratory of Organ Fibrosis Prophylaxis and Treatment by Combine Traditional Chinese and Western Medicine, Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Li Wang
- Laboratory of Organ Fibrosis Prophylaxis and Treatment by Combine Traditional Chinese and Western Medicine, Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China,
| |
Collapse
|
42
|
IL-17A as a Potential Therapeutic Target for Patients on Peritoneal Dialysis. Biomolecules 2020; 10:biom10101361. [PMID: 32987705 PMCID: PMC7598617 DOI: 10.3390/biom10101361] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/16/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease (CKD) is a health problem reaching epidemic proportions. There is no cure for CKD, and patients may progress to end-stage renal disease (ESRD). Peritoneal dialysis (PD) is a current replacement therapy option for ESRD patients until renal transplantation can be achieved. One important problem in long-term PD patients is peritoneal membrane failure. The mechanisms involved in peritoneal damage include activation of the inflammatory and immune responses, associated with submesothelial immune infiltrates, angiogenesis, loss of the mesothelial layer due to cell death and mesothelial to mesenchymal transition, and collagen accumulation in the submesothelial compact zone. These processes lead to fibrosis and loss of peritoneal membrane function. Peritoneal inflammation and membrane failure are strongly associated with additional problems in PD patients, mainly with a very high risk of cardiovascular disease. Among the inflammatory mediators involved in peritoneal damage, cytokine IL-17A has recently been proposed as a potential therapeutic target for chronic inflammatory diseases, including CKD. Although IL-17A is the hallmark cytokine of Th17 immune cells, many other cells can also produce or secrete IL-17A. In the peritoneum of PD patients, IL-17A-secreting cells comprise Th17 cells, γδ T cells, mast cells, and neutrophils. Experimental studies demonstrated that IL-17A blockade ameliorated peritoneal damage caused by exposure to PD fluids. This article provides a comprehensive review of recent advances on the role of IL-17A in peritoneal membrane injury during PD and other PD-associated complications.
Collapse
|
43
|
Mehrotra P, Ullah MM, Collett JA, Myers SL, Dwinell MR, Geurts AM, Basile DP. Mutation of RORγT reveals a role for Th17 cells in both injury and recovery from renal ischemia-reperfusion injury. Am J Physiol Renal Physiol 2020; 319:F796-F808. [PMID: 32924545 DOI: 10.1152/ajprenal.00187.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
To investigate T helper type 17 (Th17) cells in the setting of acute kidney injury, the gene encoding the master regulator of Th17 cell differentiation, that is, RAR-related orphan receptor-γ (RORγT), was mutated in Lewis rats using CRISPR/Cas9 technology. In response to 40 min of bilateral renal ischemia-reperfusion (I/R), RAR-related orphan receptor C (Rorc)-/- rats were resistant to injury relative to wild-type Rorc+/+ rats. This protection was associated with inhibition of IL-17 expression and reduced infiltration of CD4+ cells, CD8+ cells, B cells, and macrophages. To evaluate the effect of Th17 cells on repair, ischemia was increased to 50 min in Rorc-/- rats. This maneuver equalized the initial level of injury in Rorc-/- and Rorc+/+ rats 1 to 2 days post-I/R based on serum creatinine values. However, Rorc-/- rats, but not Rorc+/+ rats, failed to successfully recover renal function and had high mortality by 4 days post-I/R. Histological assessment of kidney tubules showed evidence of repair by day 4 post-I/R in Rorc+/+ rats but persistent necrosis and elevated cell proliferation in Rorc-/- rats. Adoptive transfer of CD4+ cells from the spleen of Rorc+/+ rats or supplementation of exogenous rIL-17 by an osmotic minipump improved renal function and survival of Rorc-/- rats following 50 min of I/R. This was associated with a relative decrease in the number of M1-type macrophages and a relative increase in the percentage of T regulatory cells. Taken together, these data suggest that Th17 cells have both a deleterious and a beneficial role in kidney injury and recovery, contributing to early postischemic injury and inflammation but also possibly being critical in the resolution of inflammation during kidney repair.
Collapse
Affiliation(s)
- Purvi Mehrotra
- Department of Anatomy, Cell Biology and Physiology, Indiana University of Medicine, Indianapolis, Indiana
| | - Md Mahbub Ullah
- Department of Anatomy, Cell Biology and Physiology, Indiana University of Medicine, Indianapolis, Indiana
| | - Jason A Collett
- Department of Anatomy, Cell Biology and Physiology, Indiana University of Medicine, Indianapolis, Indiana
| | - Sarah L Myers
- Department of Anatomy, Cell Biology and Physiology, Indiana University of Medicine, Indianapolis, Indiana
| | - Melinda R Dwinell
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Aron M Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - David P Basile
- Department of Anatomy, Cell Biology and Physiology, Indiana University of Medicine, Indianapolis, Indiana
| |
Collapse
|
44
|
Ullah MM, Basile DP. Role of Renal Hypoxia in the Progression From Acute Kidney Injury to Chronic Kidney Disease. Semin Nephrol 2020; 39:567-580. [PMID: 31836039 DOI: 10.1016/j.semnephrol.2019.10.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Over the past 20 years, there has been an increased appreciation of the long-term sequelae of acute kidney injury (AKI) and the potential development of chronic kidney disease (CKD). Several pathophysiologic features have been proposed to mediate AKI to CKD progression including maladaptive alterations in tubular, interstitial, inflammatory, and vascular cells. These alterations likely interact to culminate in the progression to CKD. In this article we focus primarily on evidence of vascular rarefaction secondary to AKI, and the potential mechanisms by which rarefaction occurs in relation to other alterations in tubular and interstitial compartments. We further focus on the potential that rarefaction contributes to renal hypoxia. Consideration of the role of hypoxia in AKI to CKD transition focuses on experimental evidence of persistent renal hypoxia after AKI and experimental maneuvers to evaluate the influence of hypoxia, per se, in progressive disease. Finally, consideration of methods to evaluate hypoxia in patients is provided with the suggestion that noninvasive measurement of renal hypoxia may provide insight into progression in post-AKI patients.
Collapse
Affiliation(s)
- Md Mahbub Ullah
- Department of Anatomy, Cell Biology and Physiology, Indiana University, Indianapolis, IN
| | - David P Basile
- Department of Medicine, Division of Nephrology, Indiana University, Indianapolis, IN.
| |
Collapse
|
45
|
Huang E, Peng N, Xiao F, Hu D, Wang X, Lu L. The Roles of Immune Cells in the Pathogenesis of Fibrosis. Int J Mol Sci 2020; 21:E5203. [PMID: 32708044 PMCID: PMC7432671 DOI: 10.3390/ijms21155203] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/19/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022] Open
Abstract
Tissue injury and inflammatory response trigger the development of fibrosis in various diseases. It has been recognized that both innate and adaptive immune cells are important players with multifaceted functions in fibrogenesis. The activated immune cells produce various cytokines, modulate the differentiation and functions of myofibroblasts via diverse molecular mechanisms, and regulate fibrotic development. The immune cells exhibit differential functions during different stages of fibrotic diseases. In this review, we summarized recent advances in understanding the roles of immune cells in regulating fibrotic development and immune-based therapies in different disorders and discuss the underlying molecular mechanisms with a focus on mTOR and JAK-STAT signaling pathways.
Collapse
Affiliation(s)
- Enyu Huang
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China; (E.H.); (F.X.)
| | - Na Peng
- Department of Rheumatology and Immunology, the Second People’s Hospital of Three Gorges University, Yichang 443000, China; (N.P.); (D.H.)
| | - Fan Xiao
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China; (E.H.); (F.X.)
| | - Dajun Hu
- Department of Rheumatology and Immunology, the Second People’s Hospital of Three Gorges University, Yichang 443000, China; (N.P.); (D.H.)
| | - Xiaohui Wang
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China; (E.H.); (F.X.)
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China; (E.H.); (F.X.)
| |
Collapse
|
46
|
Interleukin-17A induces vascular remodeling of small arteries and blood pressure elevation. Clin Sci (Lond) 2020; 134:513-527. [PMID: 32104886 DOI: 10.1042/cs20190682] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 02/25/2020] [Accepted: 02/27/2020] [Indexed: 01/20/2023]
Abstract
An important link exists between hypertension and inflammation. Hypertensive patients present elevated circulating levels of proinflammatory cytokines, including interleukin-17A (IL-17A). This cytokine participates in host defense, autoimmune and chronic inflammatory pathologies, and cardiovascular diseases, mainly through the regulation of proinflammatory factors. Emerging evidence also suggests that IL-17A could play a role in regulating blood pressure and end-organ damage. Here, our preclinical studies in a murine model of systemic IL-17A administration showed that increased levels of circulating IL-17A raised blood pressure induced inward remodeling of small mesenteric arteries (SMAs) and arterial stiffness. In IL-17A-infused mice, treatment with hydralazine and hydrochlorothiazide diminished blood pressure elevation, without modifying mechanical and structural properties of SMA, suggesting a direct vascular effect of IL-17A. The mechanisms of IL-17A seem to involve an induction of vascular smooth muscle cell (VSMC) hypertrophy and phenotype changes, in the absence of extracellular matrix (ECM) proteins accumulation. Accordingly, treatment with an IL-17A neutralizing antibody diminished SMA remodeling in a model of angiotensin II (Ang II) infusion. Moreover, in vitro studies in VSMCs reported here, provide further evidence of the direct effects of IL-17A on cell growth responses. Our experimental data suggest that IL-17A is a key mediator of vascular remodeling of the small arteries, which might contribute, at least in part, to blood pressure elevation.
Collapse
|
47
|
Dellepiane S, Leventhal JS, Cravedi P. T Cells and Acute Kidney Injury: A Two-Way Relationship. Front Immunol 2020; 11:1546. [PMID: 32765535 PMCID: PMC7379378 DOI: 10.3389/fimmu.2020.01546] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/11/2020] [Indexed: 12/29/2022] Open
Abstract
Acute Kidney Injury (AKI) complicates up to 10% of hospital admissions substantially increasing patient morbidity and mortality. Experimental evidence supports that AKI initiation and maintenance results from immune-mediated damage. Exogenous injury sources directly damage renal cells which produce pro-inflammatory mediators recruiting immune cells and furthering kidney injury. Many AKI studies focus on activation of innate immunity; major components include complement pathways, neutrophils, and monocytes. Recently, growing evidence emphasizes T lymphocytes role in affecting AKI pathogenesis and magnitude. In particular, T helper 17 lymphocytes enhance tissue injury by recruiting neutrophils and other inflammatory cells, while regulatory T cells conversely reduce renal injury and facilitate repair. Intriguingly, evidence supports local parenchymal-T cell interactions as essential to producing T cell phenotypic changes affecting long-term kidney and patient survival. Herein, we review T cells effects on AKI and patient outcomes and discuss related new therapeutic approaches to improve outcomes of affected individuals.
Collapse
Affiliation(s)
- Sergio Dellepiane
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jeremy S Leventhal
- Division of Nephrology, White Plains Hospital, White Plains, NY, United States
| | - Paolo Cravedi
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
48
|
AKI: an increasingly recognized risk factor for CKD development and progression. J Nephrol 2020; 33:1171-1187. [PMID: 32651850 DOI: 10.1007/s40620-020-00793-2] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/26/2020] [Indexed: 02/07/2023]
Abstract
Acute kidney injury (AKI) is an increasing health burden with high morbidity and mortality rates worldwide. AKI is a risk factor for chronic kidney disease (CKD) development and progression to end stage renal disease (ESRD). Rapid action is required to find treatment options for AKI, plus to anticipate the development of CKD and other complications. Therefore, it is essential to understand the pathophysiology of AKI to CKD transition. Over the last several years, research has revealed maladaptive repair to be an interplay of cell death, endothelial dysfunction, tubular epithelial cell senescence, inflammatory processes and more-terminating in fibrosis. Various pathological mechanisms have been discovered and reveal targets for potential interventions. Furthermore, there have been clinical efforts measures for AKI prevention and progression including the development of novel biomarkers and prediction models. In this review, we provide an overview of pathophysiological mechanisms involved in kidney fibrosis. Furthermore, we discuss research gaps and promising therapeutic approaches for AKI to CKD progression.
Collapse
|
49
|
Tee BC, Sun Z. Xenogeneic mesenchymal stem cell transplantation for mandibular defect regeneration. Xenotransplantation 2020; 27:e12625. [PMID: 32629548 DOI: 10.1111/xen.12625] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 03/18/2020] [Accepted: 06/06/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND It is commonly accepted that xenogeneic stem cell transplantation for tissue engineering is faced with host immune rejection. Using a rat critical-size mandibular defect model, this study examined whether the immune rejection can be evaded by diminishing T-cell immunity. METHODS To examine donor cell survival and host immune reaction, pig bone marrow-derived mesenchymal stem cells (BM-MSCs) were labeled with CM-DiI, loaded onto gelatin sponge (5 × 106 cells/scaffold), and transplanted into 5-mm mandibular defects of immunocompetent and T cell-deficient athymic rats. To examine the effects of xenogeneic BM-MSCs on bone regeneration, athymic rats undergone the same surgeries were terminated at post-operative weeks 1, 3, and 6. Control rats underwent the same jaw surgery without BM-MSC transplantation. RESULTS The density of CM-DiI-labeled BM-MSCs decreased with time in both strains of rats. Although it was substantially higher in athymic rats than in immunocompetent rats at post-operative day 1, by day 3-7 the density became comparable between the two strains of rats. Apoptosis reflected by cleaved Caspase-3 staining was low in both strains. Stronger infiltration of neutrophils, macrophages, B cells and CD8+ T cells was found in MSC-treated animals. In athymic rats, infiltration of neutrophils and macrophages was strong, but it occurred later than that in immunocompetent rats. While bone volume fraction significantly increased with time (P < .001), no difference was found between MSC-treated and control groups. CONCLUSIONS Even in hosts with deficient T-cell immunity, xenogeneic BM-MSC transplantation into mandibular critical-sized defects still faces challenges from host innate immunity, which compromises their regenerative efficacy.
Collapse
Affiliation(s)
- Boon Ching Tee
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Zongyang Sun
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
50
|
Mehrotra P, Sturek M, Neyra JA, Basile DP. Calcium channel Orai1 promotes lymphocyte IL-17 expression and progressive kidney injury. J Clin Invest 2020; 129:4951-4961. [PMID: 31415242 DOI: 10.1172/jci126108] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 08/13/2019] [Indexed: 12/21/2022] Open
Abstract
We hypothesized that the store-operated calcium entry (SOCE) channel, Orai1, participates in the activation of Th17 cells and influences renal injury. In rats, following renal ischemia/reperfusion (I/R), there was a rapid and sustained influx of Orai1+ CD4 T cells and IL-17 expression was restricted to Orai1+ cells. When kidney CD4+ cells of post-acute kidney injury (post-AKI) rats were stimulated with angiotensin II and elevated Na+ (10-7 M/170 mM) in vitro, there was an enhanced response in intracellular Ca2+ and IL-17 expression, which was blocked by SOCE inhibitors 2APB, YM58483/BTP2, or AnCoA4. In vivo, YM58483/BTP2 (1 mg/kg) attenuated IL-17+ cell activation, inflammation, and severity of AKI following either I/R or intramuscular glycerol injection. Rats treated with high-salt diet (5-9 weeks after I/R) manifested progressive disease indicated by enhanced inflammation, fibrosis, and impaired renal function. These responses were significantly attenuated by YM58483/BTP2. In peripheral blood of critically ill patients, Orai1+ cells were significantly elevated by approximately 10-fold and Th17 cells were elevated by approximately 4-fold in AKI versus non-AKI patients. Further, in vitro stimulation of CD4+ cells from AKI patients increased IL-17, which was blocked by SOCE inhibitors. These data suggest that Orai1 SOCE is a potential therapeutic target in AKI and CKD progression.
Collapse
Affiliation(s)
- Purvi Mehrotra
- Department of Anatomy, Cell Biology & Physiology, Indiana University, Indianapolis, Indiana, USA
| | - Michael Sturek
- Department of Anatomy, Cell Biology & Physiology, Indiana University, Indianapolis, Indiana, USA
| | - Javier A Neyra
- Department of Medicine, Division of Nephrology, Bone and Mineral Metabolism, University of Kentucky, Lexington, Kentucky, USA
| | - David P Basile
- Department of Anatomy, Cell Biology & Physiology, Indiana University, Indianapolis, Indiana, USA.,Department of Medicine Division of Nephrology, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|