Your browser doesn't support javascript.
loading
Tubular Epithelial Cell HMGB1 Promotes AKI-CKD Transition by Sensitizing Cycling Tubular Cells to Oxidative Stress: A Rationale for Targeting HMGB1 during AKI Recovery.
Zhao, Zhi Bo; Marschner, Julian A; Iwakura, Takamasa; Li, Chenyu; Motrapu, Manga; Kuang, Meisi; Popper, Bastian; Linkermann, Andreas; Klocke, Jan; Enghard, Philipp; Muto, Yoshiharu; Humphreys, Benjamin D; Harris, Helena Erlandsson; Romagnani, Paola; Anders, Hans-Joachim.
Afiliação
  • Zhao ZB; Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Munich, Germany.
  • Marschner JA; Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Munich, Germany.
  • Iwakura T; Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Munich, Germany.
  • Li C; Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Munich, Germany.
  • Motrapu M; Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Munich, Germany.
  • Kuang M; Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Munich, Germany.
  • Popper B; Biomedical Center, Core Facility Animal Models, LMU München, Munich, Germany.
  • Linkermann A; Division of Nephrology, Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany.
  • Klocke J; Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
  • Enghard P; Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
  • Muto Y; Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri.
  • Humphreys BD; Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri.
  • Harris HE; Department of Developmental Biology, Washington University in St. Louis, St. Louis, Missouri.
  • Romagnani P; Departments of Rheumatology and of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
  • Anders HJ; Department of Experimental and Biomedical Sciences "Mario Serio" and Nephrology and Dialysis Unit, Meyer Children's University Hospital, Florence, Italy.
J Am Soc Nephrol ; 34(3): 394-411, 2023 03 01.
Article em En | MEDLINE | ID: mdl-36857499
ABSTRACT
SIGNIFICANCE STATEMENT Cells undergoing necrosis release extracellular high mobility group box (HMGB)-1, which triggers sterile inflammation upon AKI in mice. Neither deletion of HMGB1 from tubular epithelial cells, nor HMGB1 antagonism with small molecules, affects initial ischemic tubular necrosis and immediate GFR loss upon unilateral ischemia/reperfusion injury (IRI). On the contrary, tubular cell-specific HMGB1 deficiency, and even late-onset pharmacological HMGB1 inhibition, increased functional and structural recovery from AKI, indicating that intracellular HMGB1 partially counters the effects of extracellular HMGB1. In vitro studies indicate that intracellular HMGB1 decreases resilience of tubular cells from prolonged ischemic stress, as in unilateral IRI. Intracellular HMGB1 is a potential target to enhance kidney regeneration and to improve long-term prognosis in AKI.

BACKGROUND:

Late diagnosis is a hurdle for treatment of AKI, but targeting AKI-CKD transition may improve outcomes. High mobility group box-1 (HMGB1) is a nuclear regulator of transcription and a driver of necroinflammation in AKI. We hypothesized that HMGB1 would also modulate AKI-CKD transition in other ways.

METHODS:

We conducted single-cell transcriptome analysis of human and mouse AKI and mouse in vivo and in vitro studies with tubular cell-specific depletion of Hmgb1 and HMGB1 antagonists.

RESULTS:

HMGB1 was ubiquitously expressed in kidney cells. Preemptive HMGB1 antagonism with glycyrrhizic acid (Gly) and ethyl pyruvate (EP) did not affect postischemic AKI but attenuated AKI-CKD transition in a model of persistent kidney hypoxia. Consistently, tubular Hmgb1 depletion in Pax8 rtTA, TetO Cre, Hmgb1fl/fl mice did not protect from AKI, but from AKI-CKD transition. In vitro studies confirmed that absence of HMGB1 or HMGB1 inhibition with Gly and EP does not affect ischemic necrosis of growth-arrested differentiated tubular cells but increased the resilience of cycling tubular cells that survived the acute injury to oxidative stress. This effect persisted when neutralizing extracellular HMGB1 with 2G7. Consistently, late-onset HMGB1 blockade with EP started after the peak of ischemic AKI in mice prevented AKI-CKD transition, even when 2G7 blocked extracellular HMGB1.

CONCLUSION:

Treatment of AKI could become feasible when ( 1 ) focusing on long-term outcomes of AKI; ( 2 ) targeting AKI-CKD transition with drugs initiated after the AKI peak; and ( 3 ) targeting with drugs that block HMGB1 in intracellular and extracellular compartments.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Proteína HMGB1 / Insuficiência Renal Crônica / Injúria Renal Aguda Limite: Animals / Humans Idioma: En Revista: J Am Soc Nephrol Assunto da revista: NEFROLOGIA Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Alemanha

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Proteína HMGB1 / Insuficiência Renal Crônica / Injúria Renal Aguda Limite: Animals / Humans Idioma: En Revista: J Am Soc Nephrol Assunto da revista: NEFROLOGIA Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Alemanha