Trends in Cell Biology
Volume 32, Issue 10, October 2022, Pages 841-853
Journal home page for Trends in Cell Biology

Review
The multifaceted role of kidney tubule mitochondrial dysfunction in kidney disease development

https://doi.org/10.1016/j.tcb.2022.03.012Get rights and content

Highlights

  • Kidney function genetic studies and follow-up omics functional analysis highlight the key role of kidney proximal tubules and metabolism in kidney disease development.

  • The kidneys have the second-highest mitochondrial density in the body to enable the active reabsorption of nutrients and electrolytes.

  • Altered redox balance, impaired cellular energetics, increased cell death, and inflammation are observed as consequences of mitochondrial dysfunction, causing kidney disease development.

More than 800 million people suffer from kidney disease. Genetic studies and follow-up animal models and cell biological experiments indicate the key role of proximal tubule metabolism. Kidneys have one of the highest mitochondrial densities. Mitochondrial biogenesis, mitochondrial fusion and fission, and mitochondrial recycling, such as mitophagy are critical for proper mitochondrial function. Mitochondrial dysfunction can lead to an energetic crisis, orchestrate different types of cell death (apoptosis, necroptosis, pyroptosis, and ferroptosis), and influence cellular calcium levels and redox status. Collectively, mitochondrial defects in renal tubules contribute to epithelial atrophy, inflammation, or cell death, orchestrating kidney disease development.

Section snippets

The key contribution of proximal tubule metabolism to kidney disease development

The kidney maintains electrolyte and fluid balance and secretes hormones. More than 800 million people suffer from kidney disease. Kidney dysfunction will cause toxin, fluid, and electrolyte build-up. Without treatment, kidney disease can progress to end-stage kidney failure requiring life-sustaining renal replacement therapy. New drug development for kidney disease is limited by our poor mechanistic understanding of disease pathogenesis. Kidney function genetic studies have highlighted

Energy balance

The primary function of the mitochondria is to generate ATP via a chain of biochemical reactions called the Krebs cycle [7]. Kidney tubules, especially proximal tubules transport kilograms of sodium chloride, other electrolytes, and nutrients daily. Kidney tubule cells preferentially oxidase fatty acids to generate energy (Figure 1). It has been known that kidney tubules can also burn ketones and lactate. While glucose utilization is almost undetectable in proximal renal tubules, it could be

Redox regulation and oxidative stress

Defective mitochondria fail to maintain the proton gradient across the inner mitochondrial membrane and are the main source of ROS in most cells (Figure 2). Under physiological conditions, 0.2–2% of the electrons in the electron transport chain do not follow the normal transfer but directly leak out of the electron transport chain and interact with oxygen to produce superoxide or hydrogen peroxide. Complexes I and III are considered to be the main sites for ROS production. In addition, NADPH

Mitochondria biogenesis

Mitochondrial numbers are a key determinant of mitochondrial function and the key mechanism to adapt to higher energy demand. Mitochondrial biogenesis is an important determinant of mitochondrial number. PGC1a is a master transcriptional regulator of mitochondrial biogenesis. Expression of PGC1a is lower in kidneys of patients with AKI and CKD [10]. Mice with genetic deletion of PCG1a appear healthy at baseline but show increased susceptibility to acute and chronic kidney injury [28]. At the

Mitochondria dynamics, shape, size, and turnover

Mitochondrial shape and size, which are controlled by fission and fusion, are other less well-understood determinants of function. Abnormalities in mitochondrial dynamics have been observed both in acute and chronic disease conditions [77]. Dynamin-related protein (DRP)1 is the master regulator of mitochondrial fission. Proximal-tubule-specific deletion of DRP1 or mdivi-1 treatment, a pharmacological inhibitor of DRP1, preserves mitochondrial structure, reduces oxidative stress, and protects

The role of mitochondria in controlling inflammation

Severe mitochondrial damage and defects in mitochondrial clearance can lead to a leakage of mtDNA into the cytosol. The exact mechanism of the mtDNA leakage is still not fully understood (Figure 4). BAK and BAX pores in the mitochondrial outer membrane can lead to inner mitochondrial membrane herniation and cytosolic release of mtDNA [6]. The presence of mtDNA in the cytosol is a sign of pathogenic infection and is recognized by the cytosolic nucleotide-sensing pathways. The nucleic acid

Apoptosis

The mitochondria play a key role in orchestrating a multitude of cell death mechanisms. Apoptosis is a noninflammatory cell death mechanism occurring without the rupture of the cell membrane. The release of cytochrome c from the mitochondria to the cytosol through BAX/BAK pores activates subsequent caspases such as caspase-9, followed by the activation of the execution caspases such as caspase-3 (Figure 5). Apoptosis has been observed in AKI and CKD and is likely a leading mechanism resulting

Concluding remarks

The kidney tubules have one of the highest mitochondrial densities to generate energy for the transport of large amounts of sodium and other solutes. Defects in mitochondrial biogenesis, dynamics, and mitophagy contribute to kidney disease development by the cells failing to meet the cellular energetic requirement. Mitochondrial damage has been widely recognized in AKI or CKD. Mitochondrial injury triggers multiple cell death mechanisms (apoptosis, necroptosis, pyroptosis, and ferroptosis)

Acknowledgments

This work was supported in the Susztak laboratory by the National Institute of Health NIH R01 DK087635, DK076077.

Declaration of Interests

The authors declare no competing interests.

References (111)

  • A. Belavgeni

    Ferroptosis and necroptosis in the kidney

    Cell Chem. Biol.

    (2020)
  • X.M. Meng

    NADPH oxidase 4 promotes cisplatin-induced acute kidney injury via ROS-mediated programmed cell death and inflammation

    Lab. Investig.

    (2018)
  • N. Miao

    The cleavage of gasdermin D by caspase-11 promotes tubular epithelial cell pyroptosis and urinary IL-18 excretion in acute kidney injury

    Kidney Int.

    (2019)
  • Z. Zhao

    XJB-5-131 inhibited ferroptosis in tubular epithelial cells after ischemia-reperfusion injury

    Cell Death Dis.

    (2020)
  • A. Picca et al.

    Regulation of mitochondrial biogenesis through TFAM-mitochondrial DNA interactions: useful insights from aging and calorie restriction studies

    Mitochondrion

    (2015)
  • K.W. Chung

    Mitochondrial damage and activation of the STING pathway lead to renal inflammation and fibrosis

    Cell Metab.

    (2019)
  • K. Tsushida

    Estrogen-related receptor α is essential for maintaining mitochondrial integrity in cisplatin-induced acute kidney injury

    Biochem. Biophys. Res. Commun.

    (2018)
  • M. Zhan

    Mitochondrial dynamics: regulatory mechanisms and emerging role in renal pathophysiology

    Kidney Int.

    (2013)
  • Y. Wang

    The PINK1/PARK2/optineurin pathway of mitophagy is activated for protection in septic acute kidney injury

    Redox Biol.

    (2021)
  • S. Li

    Drp1-regulated PARK2-dependent mitophagy protects against renal fibrosis in unilateral ureteral obstruction

    Free Radic. Biol. Med.

    (2020)
  • Z.J. Fu

    HIF-1α-BNIP3-mediated mitophagy in tubular cells protects against renal ischemia/reperfusion injury

    Redox Biol.

    (2020)
  • H. Zhang

    Mitochondrial autophagy is an HIF-1-dependent adaptive metabolic response to hypoxia

    J. Biol. Chem.

    (2008)
  • Y. Wei

    Prohibitin 2 is an inner mitochondrial membrane mitophagy receptor

    Cell

    (2017)
  • H. Maekawa

    Mitochondrial damage causes inflammation via cGAS-STING signaling in acute kidney injury

    Cell Rep.

    (2019)
  • J. Wu

    APOL1 risk variants in individuals of African genetic ancestry drive endothelial cell defects that exacerbate sepsis

    Immunity

    (2021)
  • J.F. O'Toole

    Renal manifestations of genetic mitochondrial disease

    Int. J. Nephrol. Renov. Dis.

    (2014)
  • P. Bhargava et al.

    Mitochondrial energetics in the kidney

    Nat. Rev. Nephrol.

    (2017)
  • Z. Tian et al.

    Renal metabolism and hypertension

    Nat. Commun.

    (2021)
  • A. Linkermann

    Regulated cell death in AKI

    J. Am. Soc. Nephrol.

    (2014)
  • K. McArthur

    BAK/BAX macropores facilitate mitochondrial herniation and mtDNA efflux during apoptosis

    Science

    (2018)
  • P. Mishra et al.

    Metabolic regulation of mitochondrial dynamics

    J. Cell Biol.

    (2016)
  • H.M. Kang

    Defective fatty acid oxidation in renal tubular epithelial cells has a key role in kidney fibrosis development

    Nat. Med.

    (2015)
  • V. Miguel

    Renal tubule Cpt1a overexpression protects from kidney fibrosis by restoring mitochondrial homeostasis

    J. Clin. Invest.

    (2021)
  • K.W. Chung

    Impairment of PPARα and the fatty acid oxidation pathway aggravates renal fibrosis during aging

    J. Am. Soc. Nephrol.

    (2018)
  • R. Lakhia

    PPARα agonist fenofibrate enhances fatty acid β-oxidation and attenuates polycystic kidney and liver disease in mice

    Am. J. Physiol. Ren. Physiol.

    (2018)
  • R.L. Attridge

    Fenofibrate-associated nephrotoxicity: a review of current evidence

    Am. J. Health Syst. Pharm.

    (2013)
  • M.D. Hirschey

    SIRT3 regulates mitochondrial fatty-acid oxidation by reversible enzyme deacetylation

    Nature

    (2010)
  • R. Tao

    Regulation of MnSOD enzymatic activity by Sirt3 connects the mitochondrial acetylome signaling networks to aging and carcinogenesis

    Antioxid. Redox Signal.

    (2014)
  • M. Morigi

    Sirtuin 3-dependent mitochondrial dynamic improvements protect against acute kidney injury

    J. Clin. Invest.

    (2015)
  • T.N. Haschler

    Sirtuin 5 depletion impairs mitochondrial function in human proximal tubular epithelial cells

    Sci. Rep.

    (2021)
  • T. Chiba

    Sirtuin 5 regulates proximal tubule fatty acid oxidation to protect against AKI

    J. Am. Soc. Nephrol.

    (2019)
  • J.M. Weinberg

    Mitochondrial biogenesis in kidney disease

    J. Am. Soc. Nephrol.

    (2011)
  • A. Poyan Mehr

    De novo NAD(+) biosynthetic impairment in acute kidney injury in humans

    Nat. Med.

    (2018)
  • M. Bugarski

    Changes in NAD and lipid metabolism drive acidosis-induced acute kidney injury

    J. Am. Soc. Nephrol.

    (2021)
  • M. Zheng

    Nicotinamide reduces renal interstitial fibrosis by suppressing tubular injury and inflammation

    J. Cell. Mol. Med.

    (2019)
  • Y. Guan

    Nicotinamide mononucleotide, an NAD(+) precursor, rescues age-associated susceptibility to AKI in a sirtuin 1-dependent manner

    J. Am. Soc. Nephrol.

    (2017)
  • M.T. Tran

    PGC1α drives NAD biosynthesis linking oxidative metabolism to renal protection

    Nature

    (2016)
  • V.H. Haase

    Mechanisms of hypoxia responses in renal tissue

    J. Am. Soc. Nephrol.

    (2013)
  • I. Hwang

    Catalase deficiency accelerates diabetic renal injury through peroxisomal dysfunction

    Diabetes

    (2012)
  • P. Kameritsch

    The mitochondrial thioredoxin reductase system (TrxR2) in vascular endothelium controls peroxynitrite levels and tissue integrity

    Proc. Natl. Acad. Sci. U. S. A.

    (2021)
  • Cited by (37)

    View all citing articles on Scopus
    View full text