1
|
Wu T, Zhu L, Yu M, Cai X, Chen L, Zhang H, Wu X, Ding C, Liu H, Zhang S, Li C, Shi X, Wang Y, Liu Y. AMPK Signaling Pathway Regulates Tendon Regeneration via Fatty Acid Metabolism. J Orthop Res 2025; 43:1012-1023. [PMID: 40023773 DOI: 10.1002/jor.26061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/02/2025] [Accepted: 01/28/2025] [Indexed: 03/04/2025]
Abstract
Tendon and ligament injuries are the most common musculoskeletal injuries, and their regeneration is a complex process due to the poor natural healing ability of these tissues. The current therapies for tendon repair are limited in efficacy and their cellular and molecular mechanisms remain unclear. In this study, we identified AMP-activated protein kinase (AMPK) as a markedly upregulated factor in newborn tendons with high regenerative capacity. Both in vivo and in vitro experiments demonstrated that treatment with dorsomorphin, an AMPK inhibitor, significantly decreased the tendon healing potential. Further analyses revealed that carnitine palmitoyltransferase 1A, a key enzyme, is a putative downstream target of AMPK and is closely associated with the proliferation and tenogenic differentiation of tendon stem/progenitor cells. Collectively, we highlight the essential role of AMPK in tendon repair and propose a potential therapeutic intervention for tendon injuries.
Collapse
Affiliation(s)
- Tianhao Wu
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Biomimetic Nanomaterials, Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Lisha Zhu
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
- Department of Orthodontics, School & Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, Shanghai, China
| | - Min Yu
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Biomimetic Nanomaterials, Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Xinjia Cai
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Biomimetic Nanomaterials, Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Liyuan Chen
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Biomimetic Nanomaterials, Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - He Zhang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Biomimetic Nanomaterials, Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Xiaolan Wu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Biomimetic Nanomaterials, Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Chengye Ding
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Biomimetic Nanomaterials, Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Hangbo Liu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Biomimetic Nanomaterials, Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Shiying Zhang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Biomimetic Nanomaterials, Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Chang Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Biomimetic Nanomaterials, Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Xinmeng Shi
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Biomimetic Nanomaterials, Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Yu Wang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Biomimetic Nanomaterials, Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Yan Liu
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Biomimetic Nanomaterials, Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| |
Collapse
|
2
|
Huang Z, Liao Y, Zheng Y, Ye S, Zhang Q, Yu X, Liu X, Li N. Zinc Deficiency Causes Glomerulosclerosis and Renal Interstitial Fibrosis Through Oxidative Stress and Increased Lactate Metabolism in Rats. Biol Trace Elem Res 2025; 203:2084-2098. [PMID: 39028478 PMCID: PMC11919932 DOI: 10.1007/s12011-024-04306-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/07/2024] [Indexed: 07/20/2024]
Abstract
Chronic kidney disease (CKD) is a highly prevalent condition characterized by renal fibrosis as its ultimate manifestation. Zinc deficiency is closely associated with CKD, evidenced by its link to renal fibrosis. Recently, local lactic acidosis has been demonstrated to promote renal fibrosis. Under zinc-deficient conditions, mitochondrial function is compromised and abnormal lactate metabolism might be induced potentially. However, it remains unclear whether zinc deficiency leads to renal fibrosis through local lactic acidosis. Zinc deficiency rat models were successfully established by feeding zinc-deficient diet. Western blot, qPCR, IHC, and other experiments were employed to investigate the key markers and molecular mechanisms of glomerulosclerosis and renal interstitial fibrosis. Our results indicate that zinc deficiency reduces specific markers of podocytes (podocalyxin, WT1, and nephrin) and activates the Wnt3a/β-catenin pathway, a key pathway in podocyte injury. Concurrently, glomerulosclerosis is indicated by increased urinary microalbumin and serum creatinine levels along with histological alteration observed through PAS and Masson staining in zinc-deficient rats. Furthermore, various degrees of upregulation for several markers of interstitial fibrosis including α-SMA, FN1 and collagen III are also revealed. These findings were further confirmed by Masson staining and IHC. Additionally, alterations in four markers in the EMT process, N-cadherin, E-cadherin, Vimentin, and snail, were consistent with expectations. We then confirmed the activation of the non-canonical TGF-β1 pathway known as the PI3K/AKT/mTOR pathway. An elevation in renal ROS levels accompanied by increased mitochondrial marker cytochrome C expression as well as an elevated NADH/NAD + ratio is also observed within the kidneys. Furthermore, the activity of both MMP/TIMP system and fibrinolytic system was abnormally enhanced under zinc deficiency conditions. Finally, we find zinc supplementation could significantly ameliorate relevant pathological alterations induced by zinc deficiency. These results collectively point that zinc deficiency causes podocyte damage ultimately resulting in glomerulosclerosis via accumulation of ROS and induces interstitial fibrosis via lactic acidosis.
Collapse
Affiliation(s)
- Zixuan Huang
- Department of Nephrology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Queen Mary University of London, London, E1 4NS, UK
| | - Yajie Liao
- Queen Mary University of London, London, E1 4NS, UK
| | - Yunxi Zheng
- Queen Mary University of London, London, E1 4NS, UK
| | - Shang Ye
- Queen Mary University of London, London, E1 4NS, UK
| | - Qianyu Zhang
- Queen Mary University of London, London, E1 4NS, UK
| | - Xiaohong Yu
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Xiaoxin Liu
- Department of Nephrology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Ningxu Li
- Department of Nephrology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
3
|
Aranda-Rivera AK, Amador-Martínez I, Aparicio-Trejo OE, León-Contreras JC, Hernández-Pando R, Saavedra E, García-Arroyo FE, Pedraza-Chaverri J, Sánchez-Lozada LG, Tapia E. Sulforaphane Restores Mitochondrial β-Oxidation and Reduces Renal Lipid Accumulation in a Model of Releasing Unilateral Ureteral Obstruction. Antioxidants (Basel) 2025; 14:288. [PMID: 40227243 PMCID: PMC11939561 DOI: 10.3390/antiox14030288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/11/2025] [Accepted: 02/26/2025] [Indexed: 04/15/2025] Open
Abstract
Obstructive nephropathy (ON), characterized by urine flow disruption, can induce chronic kidney disease (CKD). Although the release of the obstruction is performed as the primary intervention, renal pathology often persists and progresses. Accordingly, the murine model of releasing unilateral ureteral obstruction (RUUO) is valuable for investigating the molecular events underlying renal damage after obstruction release. Remarkably, after RUUO, disturbances such as oxidative stress, inflammation, lipid accumulation, and fibrosis continue to increase. Mitochondrial dysfunction contributes to fibrosis in the UUO model, but its role in RUUO remains unclear. Additionally, the impact of using antioxidants to restore mitochondrial function and prevent renal fibrosis in RUUO has not been determined. This study aimed to determine the therapeutic effect of pre-administering the antioxidant sulforaphane (SFN) in the RUUO model. SFN was administered 1 day before RUUO to evaluate mitochondrial biogenesis, fatty acids (FA) metabolism, bioenergetics, dynamics, and mitophagy/autophagy mechanisms in the kidney. Our data demonstrated that SFN enhanced mitochondrial biogenesis and reestablished mitochondrial oxygen consumption and β-oxidation. These effects collectively reduced lipid accumulation and normalized mitochondrial dynamics, mitophagy, and autophagy, thereby mitigating fibrosis after obstruction. Our findings suggest that SFN holds promise as a potential therapeutic agent in ON-induced CKD progression in RUUO and opens new avenues in studying antioxidant molecules to treat this disease.
Collapse
Affiliation(s)
- Ana Karina Aranda-Rivera
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Mexico City 04510, Mexico
| | - Isabel Amador-Martínez
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Mexico City 04510, Mexico
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyocán, Mexico City 04510, Mexico
| | - Omar Emiliano Aparicio-Trejo
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Juan Carlos León-Contreras
- Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City 14080, Mexico
| | - Rogelio Hernández-Pando
- Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City 14080, Mexico
| | - Emma Saavedra
- Departamento de Bioquímica, Instituto Nacional de Cardiología “Ignacio Chávez”, Mexico City 14080, Mexico
| | - Fernando E. García-Arroyo
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - José Pedraza-Chaverri
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Mexico City 04510, Mexico
| | - Laura Gabriela Sánchez-Lozada
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Edilia Tapia
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| |
Collapse
|
4
|
de la Serna-Soto M, Calleros L, Martos-Elvira M, Moreno-Piedra A, García-Villoria S, Griera M, Alcalde-Estévez E, Asenjo-Bueno A, Rodríguez-Puyol D, de Frutos S, Ruiz-Torres MP. Integrin-Linked Kinase (ILK) Promotes Mitochondrial Dysfunction by Decreasing CPT1A Expression in a Folic Acid-Based Model of Kidney Disease. Int J Mol Sci 2025; 26:1861. [PMID: 40076489 PMCID: PMC11899702 DOI: 10.3390/ijms26051861] [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: 01/16/2025] [Revised: 02/07/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Integrin-linked kinase (ILK) is a key scaffolding protein between extracellular matrix protein and the cytoskeleton and has been implicated previously in the pathogenesis of renal damage. However, its involvement in renal mitochondrial dysfunction remains to be elucidated. We studied the role of ILK and its downstream regulations in renal damage and mitochondria function both in vivo and vitro, using a folic acid (FA)-induced kidney disease model. Wild type (WT) and ILK conditional-knockdown (cKD-ILK) mice were injected with a single intraperitoneal dose of FA and studied after 15 days of chronic renal damage progression. Human Kidney tubular epithelial cells (HK2) were transfected with specific siRNAs targeting ILK, glycogen synthase kinase 3-β (GSK3β), or CCAAT/enhancer binding protein-β (C/EBPβ). The expressions and activities of renal ILK, GSK3β, C/EBPβ, mitochondrial oxidative phosphorylation enzymes, and mitochondrial membrane potential were assessed. Additionally, the expression of markers for fibrosis fibronectin (FN) and collagen 1 (COL1A1), for autophagy p62 and cytosolic light chain 3 (LC3B) isoforms II and I, and mitochondrial homeostasis marker carnitine palmitoyl-transferase 1A (CPT1A) were evaluated using immunoblotting, RT-qPCR, immunofluorescence, or colorimetric assays. FA upregulated ILK expression, leading to the decrease of GSK3β activity, increased tubular fibrosis, and produced mitochondrial dysfunction, both in vivo and vitro. These alterations were fully or partially reversed upon ILK depletion, mitigating FA-induced renal damage. The signaling axis composed by ILK, GSK3β, and C/EBPβ regulated CPT1A transcription as the limiting factor in the FA-based impaired mitochondrial activity. We highlight ILK as a potential therapeutical target for preserving mitochondrial function in kidney injury.
Collapse
Affiliation(s)
- Mariano de la Serna-Soto
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - Laura Calleros
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - María Martos-Elvira
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - Ariadna Moreno-Piedra
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - Sergio García-Villoria
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - Mercedes Griera
- Graphenano Medical Care S.L., Alcalá de Henares, 28871 Madrid, Spain;
| | - Elena Alcalde-Estévez
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - Ana Asenjo-Bueno
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - Diego Rodríguez-Puyol
- Department of Medicine, Universidad de Alcalá, Nephrology Service at Hospital Príncipe de Asturias, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain;
| | - Sergio de Frutos
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| | - María Piedad Ruiz-Torres
- Department of Systems Biology, Universidad de Alcalá, Instituto Ramon y Cajal de Investigación Sanitaria, RICORS 2040, Fundación Renal Iñigo Álvarez de Toledo, INNOREN-CM, Alcalá de Henares, 28871 Madrid, Spain; (M.d.l.S.-S.); (L.C.); (M.M.-E.); (A.M.-P.); (S.G.-V.); (E.A.-E.); (A.A.-B.); (M.P.R.-T.)
| |
Collapse
|
5
|
Li X, Xu R, Zhang D, Cai J, Zhou H, Song T, Wang X, Kong Q, Li L, Liu Z, He Z, Tang Z, Tan J, Zhang J. Baicalin: a potential therapeutic agent for acute kidney injury and renal fibrosis. Front Pharmacol 2025; 16:1511083. [PMID: 39911847 PMCID: PMC11795133 DOI: 10.3389/fphar.2025.1511083] [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: 10/14/2024] [Accepted: 01/06/2025] [Indexed: 02/07/2025] Open
Abstract
Acute kidney injury (AKI) is a common critical clinical disease that is linked to significant morbidity, recurrence, and mortality. It is characterized by a fast and prolonged loss in renal function arising from numerous etiologies and pathogenic pathways. Renal fibrosis, defined as the excessive accumulation of collagen and proliferation of fibroblasts within renal tissues, contributes to the structural damage and functional decline of the kidneys, playing a pivotal role in the advancement of Chronic Kidney Disease (CKD). Until now, while continuous renal replacement therapy (CRRT) has been utilized in the management of severe AKI, there remains a dearth of effective targeted therapies for AKI stemming from diverse etiologies. Similarly, the identification of specific biomarkers and pharmacological targets for the treatment of renal fibrosis remains a challenge. Baicalin, a naturally occurring compound classified within the flavonoid group and commonly found in the Chinese herb Scutellaria baicalensis, has shown a range of pharmacological characteristics, such as antioxidant, anti-inflammatory, antifibrotic, antitumor and antiviral effects, as evidenced by research studies. Research shows that Baicalin has potential in treating kidney diseases like AKI and renal fibrosis. This review aims to summarize Baicalin's progress in these areas, including its molecular mechanism, application in treatment, and absorption, distribution, metabolism, and excretion. Baicalin's therapeutic effects are achieved through various pathways, including antioxidant, anti-inflammatory, antifibrosis, and regulation of apoptosis and cell proliferation. Besides, we also hope this review may give some enlightenment for treating AKI and renal fibrosis in clinical practice.
Collapse
Affiliation(s)
- Xiaoming Li
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Rui Xu
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Dan Zhang
- Zunyi Medical University Library Administrative Office, Zunyi, China
| | - Ji Cai
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - He Zhou
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Tao Song
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Xianyao Wang
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Qinghong Kong
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, China
| | - Liujin Li
- Department of Otolaryngology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhaohui Liu
- Department of Otolaryngology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhixu He
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China
| | - Zhengzhen Tang
- Department of Pediatrics, The First People’s Hospital of Zunyi, Third Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jun Tan
- Department of Histology and Embryology, Zunyi Medical University, Zunyi, China
| | - Jidong Zhang
- Department of Immunology, Zunyi Medical University, Zunyi, China
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China
| |
Collapse
|
6
|
Ma Z, Huang X, Kuang J, Wang Q, Qin Y, Huang T, Liang Z, Li W, Fu Y, Li P, Fan Y, Zhai Z, Wang X, Ming J, Zhao C, Wang B, Pei D. Cpt1a Drives primed-to-naïve pluripotency transition through lipid remodeling. Commun Biol 2024; 7:1223. [PMID: 39349670 PMCID: PMC11442460 DOI: 10.1038/s42003-024-06874-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 09/10/2024] [Indexed: 10/04/2024] Open
Abstract
Metabolism has been implicated in cell fate determination, particularly through epigenetic modifications. Similarly, lipid remodeling also plays a role in regulating cell fate. Here, we present comprehensive lipidomics analysis during BMP4-driven primed to naive pluripotency transition or BiPNT and demonstrate that lipid remodeling plays an essential role. We further identify Cpt1a as a rate-limiting factor in BiPNT, driving lipid remodeling and metabolic reprogramming while simultaneously increasing intracellular acetyl-CoA levels and enhancing H3K27ac at chromatin open sites. Perturbation of BiPNT by histone acetylation inhibitors suppresses lipid remodeling and pluripotency transition. Together, our study suggests that lipid remodeling promotes pluripotency transitions and further regulates cell fate decisions, implicating Cpt1a as a critical regulator between primed-naive cell fate control.
Collapse
Affiliation(s)
- Zhaoyi Ma
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Xingnan Huang
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Junqi Kuang
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Qiannan Wang
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Yue Qin
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Tao Huang
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Zechuan Liang
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Wei Li
- Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yu Fu
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Pengli Li
- Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yixin Fan
- Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ziwei Zhai
- Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaomin Wang
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Jin Ming
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Chengchen Zhao
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Biomedical Intelligent Computing Technology, Hangzhou, China
| | - Bo Wang
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
- Zhejiang Key Laboratory of Biomedical Intelligent Computing Technology, Hangzhou, China
- Zhejiang University of Science and Technology School of Information and Electronic Engineering, Hangzhou, China
| | - Duanqing Pei
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
7
|
Miguel V, Alcalde-Estévez E, Sirera B, Rodríguez-Pascual F, Lamas S. Metabolism and bioenergetics in the pathophysiology of organ fibrosis. Free Radic Biol Med 2024; 222:85-105. [PMID: 38838921 DOI: 10.1016/j.freeradbiomed.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/15/2024] [Accepted: 06/02/2024] [Indexed: 06/07/2024]
Abstract
Fibrosis is the tissue scarring characterized by excess deposition of extracellular matrix (ECM) proteins, mainly collagens. A fibrotic response can take place in any tissue of the body and is the result of an imbalanced reaction to inflammation and wound healing. Metabolism has emerged as a major driver of fibrotic diseases. While glycolytic shifts appear to be a key metabolic switch in activated stromal ECM-producing cells, several other cell types such as immune cells, whose functions are intricately connected to their metabolic characteristics, form a complex network of pro-fibrotic cellular crosstalk. This review purports to clarify shared and particular cellular responses and mechanisms across organs and etiologies. We discuss the impact of the cell-type specific metabolic reprogramming in fibrotic diseases in both experimental and human pathology settings, providing a rationale for new therapeutic interventions based on metabolism-targeted antifibrotic agents.
Collapse
Affiliation(s)
- Verónica Miguel
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
| | - Elena Alcalde-Estévez
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain; Department of Systems Biology, Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Belén Sirera
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain
| | - Fernando Rodríguez-Pascual
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain
| | - Santiago Lamas
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain.
| |
Collapse
|
8
|
Katsuma Y, Matsui I, Matsumoto A, Okushima H, Imai A, Sakaguchi Y, Yamamoto T, Mizui M, Uchinomiya S, Kato H, Ojida A, Takashima S, Inoue K, Isaka Y. Endogenous activation of peroxisome proliferator-activated receptor-α in proximal tubule cells in counteracting phosphate toxicity. Am J Physiol Renal Physiol 2024; 327:F208-F223. [PMID: 38870264 DOI: 10.1152/ajprenal.00046.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/17/2024] [Accepted: 05/30/2024] [Indexed: 06/15/2024] Open
Abstract
Increased dietary phosphate consumption intensifies renal phosphate burden. Several mechanisms for phosphate-induced renal tubulointerstitial fibrosis have been reported. Considering the dual nature of phosphate as both a potential renal toxin and an essential nutrient for the body, kidneys may possess inherent protective mechanisms against phosphate overload, rather than succumbing solely to injury. However, there is limited understanding of such mechanisms. To identify these mechanisms, we conducted single-cell RNA sequencing (scRNA-seq) analysis of the kidneys of control and dietary phosphate-loaded (Phos) mice at a time point when the Phos group had not yet developed tubulointerstitial fibrosis. scRNA-seq analysis identified the highest number of differentially expressed genes in the clusters belonging to proximal tubular epithelial cells (PTECs). Based on these differentially expressed genes, in silico analyses suggested that the Phos group activated peroxisome proliferator-activated receptor-α (PPAR-α) and fatty acid β-oxidation (FAO) in the PTECs. This activation was further substantiated through various experiments, including the use of an FAO activity visualization probe. Compared with wild-type mice, Ppara knockout mice exhibited exacerbated tubulointerstitial fibrosis in response to phosphate overload. Experiments conducted with cultured PTECs demonstrated that activation of the PPAR-α/FAO pathway leads to improved cellular viability under high-phosphate conditions. The Phos group mice showed a decreased serum concentration of free fatty acids, which are endogenous PPAR-α agonists. Instead, experiments using cultured PTECs revealed that phosphate directly activates the PPAR-α/FAO pathway. These findings indicate that noncanonical metabolic reprogramming via endogenous activation of the PPAR-α/FAO pathway in PTECs is essential to counteract phosphate toxicity.NEW & NOTEWORTHY This study revealed the activation of peroxisome proliferator-activated receptor-α and fatty acid β-oxidation in proximal tubular epithelial cells as an endogenous mechanism to protect the kidney from phosphate toxicity. These findings highlight noncanonical metabolic reprogramming as a potential target for suppressing phosphate toxicity in the kidneys.
Collapse
Affiliation(s)
- Yusuke Katsuma
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Isao Matsui
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Ayumi Matsumoto
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroki Okushima
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsuhiro Imai
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yusuke Sakaguchi
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takeshi Yamamoto
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masayuki Mizui
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shohei Uchinomiya
- Medical Chemistry and Chemical Biology, Department of Medicinal Sciences, Graduate School of Pharmaceutical Science, Kyushu University, Fukuoka, Japan
| | - Hisakazu Kato
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akio Ojida
- Medical Chemistry and Chemical Biology, Department of Medicinal Sciences, Graduate School of Pharmaceutical Science, Kyushu University, Fukuoka, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazunori Inoue
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
9
|
Ke Q, Xiao Y, Liu D, Shi C, Shen R, Qin S, Jiang L, Yang J, Zhou Y. PPARα/δ dual agonist H11 alleviates diabetic kidney injury by improving the metabolic disorders of tubular epithelial cells. Biochem Pharmacol 2024; 222:116076. [PMID: 38387308 DOI: 10.1016/j.bcp.2024.116076] [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: 09/26/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 02/24/2024]
Abstract
Diabetic kidney disease (DKD) is responsible for nearly half of all end-stage kidney disease and kidney failure is a major driver of mortality among patients with diabetes. To date, few safe and effective drugs are available to reverse the decline of kidney function. Kidney tubules producing energy by fatty acid metabolism are pivotal in development and deterioration of DKD. Peroxisome proliferator-activated receptors (PPARs), comprising PPARα, PPARδ and PPARγ play a senior role in the pathogenesis of DKD for their functions in glycemic control and lipid metabolism; whereas systemic activation of PPARγ causes serious side-effects in clinical settings. Compound H11 was a potent PPARα and PPARδ (PPARα/δ) dual agonist with potent and well-balanced PPARα/δ agonistic activity and a high selectivity over PPARγ. In this study, the potential therapeutic effects of compound H11 were determined in a db/db mouse model of diabetes. Expressions of PPARα and PPARδ in nuclei of tubules were markedly reduced in diabetes. Transcriptional changes of tubular cells showed that H11 was an effective PPARα/δ dual agonist taking effects both in vivo and in vitro. Systemic administration of H11 showed glucose tolerance and lipid metabolic benefits in db/db mice. Moreover, H11 treatment exerted protective effects on diabetic kidney injury. In addition to fatty acid metabolism, H11 also regulated diabetes-induced metabolic alternations of branch chain amino acid degradation and glycolysis. The present study demonstrated a crucial role of H11 in regulation of energy homeostasis and metabolism in glucose-treated tubular cells. Overall, compound H11 holds therapeutic promise for DKD.
Collapse
Affiliation(s)
- Qingqing Ke
- Center for Kidney Disease, The Second Affiliated Hospital of Nanjing Medical University, China
| | - Yu Xiao
- Center for Kidney Disease, The Second Affiliated Hospital of Nanjing Medical University, China
| | - Dandan Liu
- Center for Kidney Disease, The Second Affiliated Hospital of Nanjing Medical University, China
| | - Caifeng Shi
- Center for Kidney Disease, The Second Affiliated Hospital of Nanjing Medical University, China
| | - Rui Shen
- Center for Kidney Disease, The Second Affiliated Hospital of Nanjing Medical University, China
| | - Songyan Qin
- Center for Kidney Disease, The Second Affiliated Hospital of Nanjing Medical University, China
| | - Lei Jiang
- Center for Kidney Disease, The Second Affiliated Hospital of Nanjing Medical University, China.
| | - Junwei Yang
- Center for Kidney Disease, The Second Affiliated Hospital of Nanjing Medical University, China.
| | - Yang Zhou
- Center for Kidney Disease, The Second Affiliated Hospital of Nanjing Medical University, China.
| |
Collapse
|
10
|
Hammoud S, Ivanova A, Osaki Y, Funk S, Yang H, Viquez O, Delgado R, Lu D, Phillips Mignemi M, Tonello J, Colon S, Lantier L, Wasserman DH, Humphreys BD, Koenitzer J, Kern J, de Caestecker M, Finkel T, Fogo A, Messias N, Lodhi IJ, Gewin LS. Tubular CPT1A deletion minimally affects aging and chronic kidney injury. JCI Insight 2024; 9:e171961. [PMID: 38516886 PMCID: PMC11063933 DOI: 10.1172/jci.insight.171961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 02/14/2024] [Indexed: 03/23/2024] Open
Abstract
Kidney tubules use fatty acid oxidation (FAO) to support their high energetic requirements. Carnitine palmitoyltransferase 1A (CPT1A) is the rate-limiting enzyme for FAO, and it is necessary to transport long-chain fatty acids into mitochondria. To define the role of tubular CPT1A in aging and injury, we generated mice with tubule-specific deletion of Cpt1a (Cpt1aCKO mice), and the mice were either aged for 2 years or injured by aristolochic acid or unilateral ureteral obstruction. Surprisingly, Cpt1aCKO mice had no significant differences in kidney function or fibrosis compared with wild-type mice after aging or chronic injury. Primary tubule cells from aged Cpt1aCKO mice had a modest decrease in palmitate oxidation but retained the ability to metabolize long-chain fatty acids. Very-long-chain fatty acids, exclusively oxidized by peroxisomes, were reduced in kidneys lacking tubular CPT1A, consistent with increased peroxisomal activity. Single-nuclear RNA-Seq showed significantly increased expression of peroxisomal FAO enzymes in proximal tubules of mice lacking tubular CPT1A. These data suggest that peroxisomal FAO may compensate in the absence of CPT1A, and future genetic studies are needed to confirm the role of peroxisomal β-oxidation when mitochondrial FAO is impaired.
Collapse
Affiliation(s)
- Safaa Hammoud
- Division of Nephrology and Hypertension, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Alla Ivanova
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Yosuke Osaki
- Division of Nephrology and Hypertension, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Steven Funk
- Division of Nephrology and Hypertension, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Haichun Yang
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Olga Viquez
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Rachel Delgado
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Dongliang Lu
- Division of Endocrinology, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | - Jane Tonello
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Selene Colon
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Louise Lantier
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - David H. Wasserman
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Benjamin D. Humphreys
- Division of Nephrology and Hypertension, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jeffrey Koenitzer
- Division of Pulmonary Critical Care Medicine, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Justin Kern
- Division of Nephrology and Hypertension, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | - Toren Finkel
- Aging Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Agnes Fogo
- Division of Nephrology and Hypertension, Department of Medicine, and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Nidia Messias
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Irfan J. Lodhi
- Division of Endocrinology, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Leslie S. Gewin
- Division of Nephrology and Hypertension, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Division of Nephrology and Hypertension, Department of Medicine, and
- Department of Medicine, Veterans Affairs Hospital, St. Louis, Missouri, USA
| |
Collapse
|
11
|
Li XY, Liu JQ, Wang Y, Chen Y, Hu WH, Lv YX, Wu Y, Lv J, Tang JM, Kong D. VNS improves VSMC metabolism and arteriogenesis in infarcted hearts through m/n-AChR-Akt-SDF-1α in adult male rats. J Mol Histol 2024; 55:51-67. [PMID: 38165566 PMCID: PMC10830782 DOI: 10.1007/s10735-023-10171-4] [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: 10/16/2022] [Accepted: 10/21/2023] [Indexed: 01/04/2024]
Abstract
Vagal nerve stimulation (VNS) provides a novel therapeutic strategy for injured hearts by activating cholinergic anti-inflammatory pathways. However, little information is available on the metabolic pattern and arteriogenesis of VSMCs after MI. VNS has been shown to stimulate the expression of CPT1α, CPT1β, Glut1, Glut4 and SDF-1α in coronary VSMCs, decreasing the number of CD68-positive macrophages while increasing CD206-positive macrophages in the infarcted hearts, leading to a decrease in TNF-α and IL-1β accompanied by a reduced ratio of CD68- and CD206-positive cells, which were dramatically abolished by atropine and mecamylamine in vivo. Knockdown of SDF-1α substantially abrogated the effect of VNS on macrophagecell alteration and inflammatory factors in infarcted hearts. Mechanistically, ACh induced SDF-1α expression in VSMCs in a dose-dependent manner. Conversely, atropine, mecamylamine, and a PI3K/Akt inhibitor completely eliminated the effect of ACh on SDF-1α expression. Functionally, VNS promoted arteriogenesis and improved left ventricular performance, which could be abolished by Ad-shSDF-1α. Thus, VNS altered the VSMC metabolism pattern and arteriogenesis to repair the infarcted heart by inducing SDF-1α expression, which was associated with the m/nAChR-Akt signaling pathway.
Collapse
Affiliation(s)
- Xing-Yuan Li
- Department of Physiology, Faculty of Basic Medical Sciences, Zunyi Medicical University, Zunyi, 563006, Guizhou, People's Republic of China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Jia-Qi Liu
- Nursing College, Hubei Province Chinese Medicine Hospital, Hubei University of Traditional Chinese Medicine, Wuhan, 430065, Hubei, People's Republic of China
| | - Yan Wang
- Department of Physiology, Faculty of Basic Medical Sciences, Zunyi Medicical University, Zunyi, 563006, Guizhou, People's Republic of China
| | - Yan Chen
- Department of Physiology, Faculty of Basic Medical Sciences, Zunyi Medicical University, Zunyi, 563006, Guizhou, People's Republic of China
| | - Wen-Hui Hu
- Department of Physiology, Faculty of Basic Medical Sciences, Zunyi Medicical University, Zunyi, 563006, Guizhou, People's Republic of China
| | - Yan-Xia Lv
- Hubei Key Laboratory of Embryonic Stem Cell Research, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Yan Wu
- Hubei Key Laboratory of Embryonic Stem Cell Research, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Jing Lv
- Institute of Basic Medical Sciences, Institute of Biomedicine, Hubei University of Medicine, Hubei, 442000, People's Republic of China.
| | - Jun-Ming Tang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.
- Department of Physiology, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.
- Institute of Basic Medical Sciences, Institute of Biomedicine, Hubei University of Medicine, Hubei, 442000, People's Republic of China.
| | - Deying Kong
- Department of Physiology, Faculty of Basic Medical Sciences, Zunyi Medicical University, Zunyi, 563006, Guizhou, People's Republic of China.
| |
Collapse
|
12
|
Hu H, Li W, Hao Y, Peng Z, Zou Z, Liang W. Baicalin ameliorates renal fibrosis by upregulating CPT1α-mediated fatty acid oxidation in diabetic kidney disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155162. [PMID: 37922789 DOI: 10.1016/j.phymed.2023.155162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/21/2023] [Accepted: 10/22/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Diabetic kidney disease (DKD) is a leading cause of end-stage renal disease (ESRD). The progression of DKD is often marked by heightened renal fibrosis due to hindered fatty acid oxidation within renal tubules. Baicalin (BA), a naturally derived compound, has exhibited the potential to mitigate the advancement of DKD. Delving deeper into the precise targets and mechanisms of BA's effect on DKD is crucial. PURPOSE This study sought to elucidate the specific mechanism through which BA moderates the progression of DKD. METHODS Renal tubular tissues from diabetic (db/db) and control (db/m) mice were subjected to mRNA sequencing to discern BA's influence on DKD. Immunohistochemical staining and Western blot were employed to assess the expression of CPT1α in DKD patients and db/db and db/m mice administered with either BA (50 mg/kg/day) or a vehicle for 12 weeks. In vitro, human proximal renal tubule cells (HK-2) were treated with 40 mM high glucose or 50 μM BA. The potential inhibitory mechanism of BA on renal fibrosis in DKD was evaluated using Oil Red O staining and oxygen consumption rate (OCR) measurements. RESULTS The results demonstrated that BA notably reduced lipid accumulation and renal fibrosis in db/db mice. Moreover, mRNA sequencing pinpointed a significant downregulation of CPT1α in DKD. In vitro assays revealed that both the overexpression of CPT1α and treatment with BA exerted similar influences on mitochondrial respiration, fatty acid oxidation, and renal fibrosis levels. Given the pronounced downregulation of CPT1α in DKD patients and its substantial correlation with clinical indicators, it was evident that CPT1α could serve as a therapeutic target for BA in addressing DKD. CONCLUSION Our findings demonstrated that BA potentially enhances FAO by augmenting the expression of CPT1α, subsequently diminishing renal fibrosis in DKD. As such, CPT1α emerges as a promising therapeutic target for DKD intervention.
Collapse
Affiliation(s)
- Hongtu Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, Hubei 430060, China; Key Clinical Research Center of Kidney Disease in Hubei, 238 Jiefang Rd, Wuhan, Hubei 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weiwei Li
- Division of Nephrology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, No.158 Wuyang Avenue, Enshi City, Hubei Province, China
| | - Yiqun Hao
- Division of Nephrology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, Hubei 430060, China; Key Clinical Research Center of Kidney Disease in Hubei, 238 Jiefang Rd, Wuhan, Hubei 430060, China
| | - Zhuan Peng
- Division of Nephrology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, Hubei 430060, China; Key Clinical Research Center of Kidney Disease in Hubei, 238 Jiefang Rd, Wuhan, Hubei 430060, China
| | - Zhengping Zou
- Division of Nephrology, Qianjiang Hospital Affiliated to Renmin Hospital of Wuhan University, No. 22, Zhanghua Zhong Road, Qianjiang, Hubei 433100, China; Qianjiang Clinical Medical College, Health Science Center, Yangtze University, China.
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, Hubei 430060, China; Key Clinical Research Center of Kidney Disease in Hubei, 238 Jiefang Rd, Wuhan, Hubei 430060, China.
| |
Collapse
|
13
|
Fan Y, Yuan Y, Xiong M, Jin M, Zhang D, Yang D, Liu C, Petersen RB, Huang K, Peng A, Zheng L. Tet1 deficiency exacerbates oxidative stress in acute kidney injury by regulating superoxide dismutase. Theranostics 2023; 13:5348-5364. [PMID: 37908721 PMCID: PMC10614682 DOI: 10.7150/thno.87416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/14/2023] [Indexed: 11/02/2023] Open
Abstract
Rationale: Increased methylation of key genes has been observed in kidney diseases, suggesting that the ten-eleven translocation (Tet) methyl-cytosine dioxygenase family as well as 5mC oxidation may play important roles. As a member of the Tet family, the role of Tet1 in acute kidney injury (AKI) remains unclear. Methods: Tet1 knockout mice, with or without tempol treatment, a scavenger of reactive oxygen species (ROS), were challenged with ischemia and reperfusion (I/R) injury or unilateral ureteral obstruction (UUO) injury. RNA-sequencing, Western blotting, qRT-PCR, bisulfite sequencing, chromatin immunoprecipitation, immunohistochemical staining, and dot blot assays were performed. Results: Tet1 expression was rapidly upregulated following I/R or UUO injury. Moreover, Tet1 knockout mice showed increased renal injury and renal cell death, increased ROS accumulation, G2/M cell cycle arrest, inflammation, and fibrosis. Severe renal damage in injured Tet1 knockout mice was alleviated by tempol treatment. Mechanistically, Tet1 reduced the 5mC levels in an enzymatic activity-dependent manner on the promoters of Sod1 and Sod2 to promote their expression, thus lowering injury-induced excessive ROS and reducing I/R or UUO injury. Conclusions: Tet1 plays an important role in the development of AKI by promoting SOD expression through a DNA demethylase-dependent mechanism.
Collapse
Affiliation(s)
- Yu Fan
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China, 430072
| | - Yangmian Yuan
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China, 430072
| | - Mingrui Xiong
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China, 430030
| | - Muchuan Jin
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China, 430072
| | - Donge Zhang
- Department of Pharmacy, The Third Hospital of Wuhan and Tongren Hospital of Wuhan University, Wuhan, China, 430070
| | - Dong Yang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China, 430030
| | - Chengyu Liu
- Department of Transfusion Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China, 430030
| | - Robert B. Petersen
- Foundational Sciences, Central Michigan University College of Medicine, Mount Pleasant, MI, USA, 48858
| | - Kun Huang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China, 430030
| | - Anlin Peng
- Department of Pharmacy, The Third Hospital of Wuhan and Tongren Hospital of Wuhan University, Wuhan, China, 430070
| | - Ling Zheng
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China, 430072
| |
Collapse
|
14
|
Rahbar Saadat Y, Hosseiniyan Khatibi SM, Sani A, Zununi Vahed S, Ardalan M. Ischemic tubular injury: Oxygen-sensitive signals and metabolic reprogramming. Inflammopharmacology 2023; 31:1657-1669. [PMID: 37131045 DOI: 10.1007/s10787-023-01232-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: 07/23/2022] [Accepted: 03/21/2023] [Indexed: 05/04/2023]
Abstract
The kidneys are the most vulnerable organs to severe ischemic insult that results in cellular hypoxia under pathophysiological conditions. Large amounts of oxygen are consumed by the kidneys, mainly to produce energy for tubular reabsorption. Beyond high oxygen demand and the low oxygen supply, different other factors make kidneys vulnerable to ischemia which is deemed to be a major cause of acute kidney injury (AKI). On the other hand, kidneys are capable of sensing and responding to oxygen alternations to evade harms resulting from inadequate oxygen. The hypoxia-inducible factor (HIF) is the main conserved oxygen-sensing mechanism that maintains homeostasis under hypoxia through direct/indirect regulation of several genes that contribute to metabolic adaptation, angiogenesis, energy conservation, erythropoiesis, and so on. In response to oxygen availability, prolyl-hydroxylases (PHDs) control the HIF stability. This review focuses on the oxygen-sensing mechanisms in kidneys, particularly in proximal tubular cells (PTCs) and discusses the molecules involved in ischemic response and metabolic reprogramming. Moreover, the possible roles of non-coding RNAs (microRNAs and long non-coding RNAs) in the development of ischemic AKI are put forward.
Collapse
Affiliation(s)
| | | | - Anis Sani
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | |
Collapse
|
15
|
Calvert ND, Kirby A, Suchý M, Pallister P, Torrens AA, Burger D, Melkus G, Schieda N, Shuhendler AJ. Direct mapping of kidney function by DCE-MRI urography using a tetrazinanone organic radical contrast agent. Nat Commun 2023; 14:3965. [PMID: 37407664 DOI: 10.1038/s41467-023-39720-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023] Open
Abstract
Chronic kidney disease (CKD) and acute kidney injury (AKI) are ongoing global health burdens. Glomerular filtration rate (GFR) is the gold standard measure of kidney function, with clinical estimates providing a global assessment of kidney health without spatial information of kidney- or region-specific dysfunction. The addition of dynamic contrast enhanced magnetic resonance imaging (DCE-MRI) to the anatomical imaging already performed would yield a 'one-stop-shop' for renal assessment in cases of suspected AKI and CKD. Towards urography by DCE-MRI, we evaluated a class of nitrogen-centered organic radicals known as verdazyls, which are extremely stable even in highly reducing environments. A glucose-modified verdazyl, glucoverdazyl, provided contrast limited to kidney and bladder, affording functional kidney evaluation in mouse models of unilateral ureteral obstruction (UUO) and folic acid-induced nephropathy (FAN). Imaging outcomes correlated with histology and hematology assessing kidney dysfunction, and glucoverdazyl clearance rates were found to be a reliable surrogate measure of GFR.
Collapse
Affiliation(s)
- Nicholas D Calvert
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, 150 Louis Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| | - Alexia Kirby
- Department of Biology, University of Ottawa, 150 Louis Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| | - Mojmír Suchý
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, 150 Louis Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| | - Peter Pallister
- Department of Chemistry, Carleton University, 1125 Colonel By Dr., Ottawa, Ontario, K1S 5B6, Canada
| | - Aidan A Torrens
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, 150 Louis Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| | - Dylan Burger
- Kidney Research Center, Ottawa Hospital Research Institute, University of Ottawa, 501 Smyth Rd, Ottawa, Ontario, K1H 8L6, Canada
| | - Gerd Melkus
- Dept. Medical Imaging, The Ottawa Hospital, 501 Smyth Rd, Ottawa, Ontario, K1H 8L6, Canada
- Dept. Radiology, University of Ottawa, 501 Smyth Rd, Ottawa, Ontario, K1H 8L6, Canada
| | - Nicola Schieda
- Dept. Radiology, University of Ottawa, 501 Smyth Rd, Ottawa, Ontario, K1H 8L6, Canada
| | - Adam J Shuhendler
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, 150 Louis Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada.
- Department of Biology, University of Ottawa, 150 Louis Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada.
- University of Ottawa Heart Institute, 40 Ruskin St., Ottawa, Ontario, K1Y 4W7, Canada.
| |
Collapse
|
16
|
Miguel V, Kramann R. Metabolic reprogramming heterogeneity in chronic kidney disease. FEBS Open Bio 2023; 13:1154-1163. [PMID: 36723270 PMCID: PMC10315765 DOI: 10.1002/2211-5463.13568] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/02/2023] Open
Abstract
Fibrosis driven by excessive accumulation of extracellular matrix (ECM) is the hallmark of chronic kidney disease (CKD). Myofibroblasts, which are the cells responsible for ECM production, are activated by cross talk with injured proximal tubule and immune cells. Emerging evidence suggests that alterations in metabolism are not only a feature of but also play an influential role in the pathogenesis of renal fibrosis. The application of omics technologies to cell-tracing animal models and follow-up functional data suggest that cell-type-specific metabolic shifts have particular roles in the fibrogenic response. In this review, we cover the main metabolic reprogramming outcomes in renal fibrosis and provide a future perspective on the field of renal fibrometabolism.
Collapse
Affiliation(s)
- Verónica Miguel
- Institute of Experimental Medicine and Systems BiologyRWTH Aachen University HospitalAachenGermany
| | - Rafael Kramann
- Institute of Experimental Medicine and Systems BiologyRWTH Aachen University HospitalAachenGermany
| |
Collapse
|
17
|
Wei X, Hou Y, Long M, Jiang L, Du Y. Advances in energy metabolism in renal fibrosis. Life Sci 2022; 312:121033. [PMID: 36270427 DOI: 10.1016/j.lfs.2022.121033] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/22/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022]
Abstract
Renal fibrosis is a common pathway toward chronic kidney disease (CKD) and is the main pathological predecessor for end-stage renal disease; thus, preventing progressive CKD and renal fibrosis is essential to reducing their consequential morbidity and mortality. Emerging evidence has connected renal fibrosis to metabolic reprogramming; abnormalities in energy metabolism pathways, such as glycolysis, the tricarboxylic acid cycle, and lipid metabolism, are known to cause diseases of diverse etiologies. Cytokine interventions in affected metabolic pathways may significantly reduce the degree of fibrosis, highlighting therapeutic targets for drug development for renal fibrosis. Here, we discuss the relationship between glycolysis, lipid metabolism, mitochondrial and peroxisome dysfunction, and renal fibrosis in detail and propose that targeted therapies for specific metabolic pathways are expected to represent the next generation of treatments for renal fibrosis.
Collapse
Affiliation(s)
- Xuejiao Wei
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Yue Hou
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Mengtuan Long
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Lili Jiang
- Department of Physical Examination Center, The First Hospital of Jilin University, Changchun, China
| | - Yujun Du
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
18
|
Ji ZH, Ren WZ, Wang HQ, Gao W, Yuan B. Molecular Subtyping Based on Cuproptosis-Related Genes and Characterization of Tumor Microenvironment Infiltration in Kidney Renal Clear Cell Carcinoma. Front Oncol 2022; 12:919083. [PMID: 35875087 PMCID: PMC9299088 DOI: 10.3389/fonc.2022.919083] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/31/2022] [Indexed: 12/30/2022] Open
Abstract
The incidence of kidney renal clear cell carcinoma (KIRC) is rising worldwide, and the prognosis is poor. Cuproptosis is a new form of cell death that is dependent on and regulated by copper ions. The relationship between cuproptosis and KIRC remains unclear. In the current study, changes in cuproptosis-related genes (CRGs) in TCGA-KIRC transcriptional datasets were characterized, and the expression patterns of these genes were analyzed. We identified three main molecular subtypes and discovered that multilayer CRG changes were associated with patient clinicopathological traits, prognosis, elesclomol sensitivity, and tumor microenvironment (TME) cell infiltration characteristics. Then, a CRG score was created to predict overall survival (OS). The CRG score was found to be strongly linked to the TME. These findings may help elucidate the roles of CRGs in KIRC, potentially enhancing understanding of cuproptosis and supporting the development of more effective immunotherapy strategies.
Collapse
Affiliation(s)
- Zhong-Hao Ji
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, China.,Department of Basic Medicine, Changzhi Medical College, Changzhi, China
| | - Wen-Zhi Ren
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, China
| | - Hao-Qi Wang
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, China
| | - Wei Gao
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, China
| | - Bao Yuan
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, China
| |
Collapse
|
19
|
Tubular Mitochondrial Dysfunction, Oxidative Stress, and Progression of Chronic Kidney Disease. Antioxidants (Basel) 2022; 11:antiox11071356. [PMID: 35883847 PMCID: PMC9311633 DOI: 10.3390/antiox11071356] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 12/23/2022] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are interconnected conditions, and CKD is projected to become the fifth leading global cause of death by 2040. New therapeutic approaches are needed. Mitochondrial dysfunction and oxidative stress have emerged as drivers of kidney injury in acute and chronic settings, promoting the AKI-to-CKD transition. In this work, we review the role of mitochondrial dysfunction and oxidative stress in AKI and CKD progression and discuss novel therapeutic approaches. Specifically, evidence for mitochondrial dysfunction in diverse models of AKI (nephrotoxicity, cytokine storm, and ischemia-reperfusion injury) and CKD (diabetic kidney disease, glomerulopathies) is discussed; the clinical implications of novel information on the key role of mitochondria-related transcriptional regulators peroxisome proliferator-activated receptor gamma coactivator 1-alpha, transcription factor EB (PGC-1α, TFEB), and carnitine palmitoyl-transferase 1A (CPT1A) in kidney disease are addressed; the current status of the clinical development of therapeutic approaches targeting mitochondria are updated; and barriers to the clinical development of mitochondria-targeted interventions are discussed, including the lack of clinical diagnostic tests that allow us to categorize the baseline renal mitochondrial dysfunction/mitochondrial oxidative stress and to monitor its response to therapeutic intervention. Finally, key milestones for further research are proposed.
Collapse
|
20
|
The Therapeutic Potential of Zinc-Alpha2-Glycoprotein (AZGP1) in Fibrotic Kidney Disease. Int J Mol Sci 2022; 23:ijms23020646. [PMID: 35054830 PMCID: PMC8775758 DOI: 10.3390/ijms23020646] [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: 12/01/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 11/30/2022] Open
Abstract
Chronic kidney disease (CKD) is characterized by a long-term loss of kidney function and, in most cases, by progressive fibrosis. Zinc-alpha2-glycoprotein (AZGP1) is a secreted protein, which is expressed in many different tissues and has been associated with a variety of functions. In a previous study, we have shown in cell culture and in AZGP1 deficient mice that AZGP1 has protective anti-fibrotic effects. In the present study, we tested the therapeutic potential of an experimental increase in AZGP1 using two different strategies. (1) C57Bl/6J mice were treated systemically with recombinant AZGP1, and (2) a transgenic mouse strain was generated to overexpress AZGP1 conditionally in proximal tubular cells. Mice underwent unilateral uretic obstruction as a pro-fibrotic kidney stress model, and kidneys were examined after 14 days. Recombinant AZGP1 treatment was accompanied by better preservation of tubular integrity, reduced collagen deposition, and lower expression of injury and fibrosis markers. Weaker but similar tendencies were observed in transgenic AZGP1 overexpressing mice. Higher AZGP1 levels led to a significant reduction in stress-induced accumulation of tubular lipid droplets, which was paralleled by improved expression of key players in lipid metabolism and fatty acid oxidation. Together these data show beneficial effects of elevated AZGP1 levels in fibrotic kidney disease and highlight a novel link to tubular cell lipid metabolism, which might open up new opportunities for CKD treatment.
Collapse
|