Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Björn Hartleben is active.

Publication


Featured researches published by Björn Hartleben.


Journal of Clinical Investigation | 2010

Autophagy influences glomerular disease susceptibility and maintains podocyte homeostasis in aging mice

Björn Hartleben; Markus Gödel; Catherine Meyer-Schwesinger; Shuya Liu; Theresa Ulrich; Sven Köbler; Thorsten Wiech; Florian Grahammer; Sebastian J. Arnold; Maja T. Lindenmeyer; Clemens D. Cohen; Hermann Pavenstädt; Dontscho Kerjaschki; Noboru Mizushima; Andrey S. Shaw; Gerd Walz; Tobias B. Huber

Injury and loss of podocytes are leading factors of glomerular disease and renal failure. The postmitotic podocyte is the primary glomerular target for toxic, immune, metabolic, and oxidant stress, but little is known about how this cell type copes with stress. Recently, autophagy has been identified as a major pathway that delivers damaged proteins and organelles to lysosomes in order to maintain cellular homeostasis. Here we report that podocytes exhibit an unusually high level of constitutive autophagy. Podocyte-specific deletion of autophagy-related 5 (Atg5) led to a glomerulopathy in aging mice that was accompanied by an accumulation of oxidized and ubiquitinated proteins, ER stress, and proteinuria. These changes resulted ultimately in podocyte loss and late-onset glomerulosclerosis. Analysis of pathophysiological conditions indicated that autophagy was substantially increased in glomeruli from mice with induced proteinuria and in glomeruli from patients with acquired proteinuric diseases. Further, mice lacking Atg5 in podocytes exhibited strongly increased susceptibility to models of glomerular disease. These findings highlight the importance of induced autophagy as a key homeostatic mechanism to maintain podocyte integrity. We postulate that constitutive and induced autophagy is a major protective mechanism against podocyte aging and glomerular injury, representing a putative target to ameliorate human glomerular disease and aging-related loss of renal function.


Molecular and Cellular Biology | 2003

Nephrin and CD2AP associate with phosphoinositide 3-OH kinase and stimulate AKT-dependent signaling

Tobias B. Huber; Björn Hartleben; Jeong Kim; Miriam Schmidts; Bernhard Schermer; Alexander Keil; Lotti Egger; Rachel L. Lecha; Christoph Borner; Hermann Pavenstädt; Andrey S. Shaw; Gerd Walz; Thomas Benzing

ABSTRACT Mutations of NPHS1 or NPHS2, the genes encoding nephrin and podocin, as well as the targeted disruption of CD2-associated protein (CD2AP), lead to heavy proteinuria, suggesting that all three proteins are essential for the integrity of glomerular podocytes, the visceral glomerular epithelial cells of the kidney. It has been speculated that these proteins participate in common signaling pathways; however, it has remained unclear which signaling proteins are actually recruited by the slit diaphragm protein complex in vivo. We demonstrate that both nephrin and CD2AP interact with the p85 regulatory subunit of phosphoinositide 3-OH kinase (PI3K) in vivo, recruit PI3K to the plasma membrane, and, together with podocin, stimulate PI3K-dependent AKT signaling in podocytes. Using two-dimensional gel analysis in combination with a phosphoserine-specific antiserum, we demonstrate that the nephrin-induced AKT mediates phosphorylation of several target proteins in podocytes. One such target is Bad; its phosphorylation and inactivation by 14-3-3 protects podocytes against detachment-induced cell death, suggesting that the nephrin-CD2AP-mediated AKT activity can regulate complex biological programs. Our findings reveal a novel role for the slit diaphragm proteins nephrin, CD2AP, and podocin and demonstrate that these three proteins, in addition to their structural functions, initiate PI3K/AKT-dependent signal transduction in glomerular podocytes.


Journal of Clinical Investigation | 2011

Role of mTOR in podocyte function and diabetic nephropathy in humans and mice

Markus Gödel; Björn Hartleben; Nadja Herbach; Shuya Liu; Stefan Zschiedrich; Shun Lu; Andrea Debreczeni-Mór; Maja T. Lindenmeyer; Maria Pia Rastaldi; Götz Hartleben; Thorsten Wiech; Alessia Fornoni; Robert G. Nelson; Matthias Kretzler; Rüdiger Wanke; Hermann Pavenstädt; Dontscho Kerjaschki; Clemens D. Cohen; Michael N. Hall; Markus A. Rüegg; Ken Inoki; Gerd Walz; Tobias B. Huber

Chronic glomerular diseases, associated with renal failure and cardiovascular morbidity, represent a major health issue. However, they remain poorly understood. Here we have reported that tightly controlled mTOR activity was crucial to maintaining glomerular podocyte function, while dysregulation of mTOR facilitated glomerular diseases. Genetic deletion of mTOR complex 1 (mTORC1) in mouse podocytes induced proteinuria and progressive glomerulosclerosis. Furthermore, simultaneous deletion of both mTORC1 and mTORC2 from mouse podocytes aggravated the glomerular lesions, revealing the importance of both mTOR complexes for podocyte homeostasis. In contrast, increased mTOR activity accompanied human diabetic nephropathy, characterized by early glomerular hypertrophy and hyperfiltration. Curtailing mTORC1 signaling in mice by genetically reducing mTORC1 copy number in podocytes prevented glomerulosclerosis and significantly ameliorated the progression of glomerular disease in diabetic nephropathy. These results demonstrate the requirement for tightly balanced mTOR activity in podocyte homeostasis and suggest that mTOR inhibition can protect podocytes and prevent progressive diabetic nephropathy.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Podocytes use FcRn to clear IgG from the glomerular basement membrane

Shreeram Akilesh; Tobias B. Huber; Hui Wu; Gary X. Wang; Björn Hartleben; Jeffrey B. Kopp; Jeffrey H. Miner; Derry C. Roopenian; Emil R. Unanue; Andrey S. Shaw

The glomerular filtration barrier prevents large serum proteins from being lost into the urine. It is not known, however, why the filter does not routinely clog with large proteins that enter the glomerular basement membrane (GBM). Here, we provide evidence that an active transport mechanism exists to remove immunoglobulins that accumulate at the filtration barrier. We found that FcRn, an IgG and albumin transport receptor, is expressed in podocytes and functions to internalize IgG from the GBM. Mice lacking FcRn accumulated IgG in the GBM as they aged, and tracer studies showed delayed clearance of IgG from the kidneys of FcRn-deficient mice. Supporting a role for this pathway in disease, saturating the clearance mechanism potentiated the pathogenicity of nephrotoxic sera. These studies support the idea that podocytes play an active role in removing proteins from the GBM and suggest that genetic or acquired impairment of the clearance machinery is likely to be a common mechanism promoting glomerular diseases.


Journal of Clinical Investigation | 2006

Bigenic mouse models of focal segmental glomerulosclerosis involving pairwise interaction of CD2AP, Fyn, and synaptopodin

Tobias B. Huber; Christopher Kwoh; Hui Wu; Katsuhiko Asanuma; Markus Gödel; Björn Hartleben; Ken J. Blumer; Jeffrey H. Miner; Peter Mundel; Andrey S. Shaw

Focal segmental glomerulosclerosis (FSGS) is the most common primary glomerular diagnosis resulting in end-stage renal disease. Defects in several podocyte proteins have been implicated in the etiology of FSGS, including podocin, alpha-actinin-4, CD2-associated protein (CD2AP), and TRPC6. Despite our growing understanding of genes involved in the pathogenesis of focal segmental sclerosis, the vast majority of patients with this disease, even those with a familial linkage, lack a clear genetic diagnosis. Here, we tested whether combinations of genetic heterozygosity (bigenic heterozygosity) that alone do not result in clinical kidney disease could function together to enhance susceptibility to glomerular damage and FSGS. Combinations of Cd2ap heterozygosity and heterozygosity of either synaptopodin (Synpo) or Fyn proto-oncogene (Fyn) but not kin of IRRE like 1 (Neph1) resulted in spontaneous proteinuria and in FSGS-like glomerular damage. These genetic interactions were also reflected at a functional level, as we found that CD2AP associates with Fyn and Synpo but not with Neph1. This demonstrates that bigenic heterozygosity can lead to FSGS and suggests that combined mutations in 2 or multiple podocyte genes may be a common etiology for glomerular disease.


Autophagy | 2012

Emerging role of autophagy in kidney function, diseases and aging

Tobias B. Huber; Charles L. Edelstein; Björn Hartleben; Ken Inoki; Man Jiang; Daisuke Koya; Shinji Kume; Wilfred Lieberthal; Nicolas Pallet; Alejandro Quiroga; Kameswaran Ravichandran; Katalin Susztak; Sei Yoshida; Zheng Dong

Autophagy is a highly conserved process that degrades cellular long-lived proteins and organelles. Accumulating evidence indicates that autophagy plays a critical role in kidney maintenance, diseases and aging. Ischemic, toxic, immunological, and oxidative insults can cause an induction of autophagy in renal epithelial cells modifying the course of various kidney diseases. This review summarizes recent insights on the role of autophagy in kidney physiology and diseases alluding to possible novel intervention strategies for treating specific kidney disorders by modifying autophagy.


Journal of The American Society of Nephrology | 2011

Prorenin Receptor Is Essential for Podocyte Autophagy and Survival

Fabian Riediger; Ivo Quack; Fatimunnisa Qadri; Björn Hartleben; Joon-Keun Park; Sebastian A. Potthoff; Dennis Sohn; Gabin Sihn; Anthony Rousselle; Verena Fokuhl; Ulrike Maschke; Bettina Purfürst; Wolfgang Schneider; Lars Christian Rump; Friedrich C. Luft; Ralf Dechend; Michael Bader; Tobias B. Huber; Genevieve Nguyen; Dominik Müller

The prorenin receptor (PRR) is highly expressed in podocytes, but its role in the maintenance of podocyte function is unknown. Here we generated podocyte-specific PRR-knockout mice and found that these animals died between 2 to 3 wk after birth. Within 14 d, PRR-knockout mice developed nephrotic syndrome, albuminuria with podocyte foot-process fusion, and cytoskeletal changes. Podocyte-specific PRR deletion also led to disturbed processing of multivesicular bodies and enrichment of autophagosomal (LC3) and lysosomal (LAMP2) markers, indicating a functional block in autophagosome-lysosome fusion and an overload of the proteasomal protein-degradation machinery. In vitro, PRR knockdown and pharmacologic blockade of vacuolar H(+)-ATPases, which associate with the PRR, increased vesicular pH, led to accumulation of LC3-positive and LAMP2-positive vesicles and altered the cytoskeleton. Taken together, these results suggest that the PRR is essential for podocyte function and survival by maintaining autophagy and protein-turnover machinery. Furthermore, PRR contributes to the control of lysosomal pH, which is important for podocyte survival and cytoskeletal integrity.


Autophagy | 2012

Autophagy plays a critical role in kidney tubule maintenance, aging and ischemia-reperfusion injury

Shuya Liu; Björn Hartleben; Oliver Kretz; Thorsten Wiech; Peter Igarashi; Noboru Mizushima; Gerd Walz; Tobias B. Huber

Autophagy is responsible for the degradation of protein aggregates and damaged organelles. Several studies have reported increased autophagic activity in tubular cells after kidney injury. Here, we examine the role of tubular cell autophagy in vivo under both physiological conditions and stress using two different tubular-specific Atg5-knockout mouse models. While Atg5 deletion in distal tubule cells does not cause a significant alteration in kidney function, deleting Atg5 in both distal and proximal tubule cells results in impaired kidney function. Already under physiological conditions, Atg5-null tubule cells display a significant accumulation of p62 and oxidative stress markers. Strikingly, tubular cell Atg5-deficiency dramatically sensitizes the kidneys to ischemic injury, resulting in impaired kidney function, accumulation of damaged mitochondria as well as increased tubular cell apoptosis and proliferation, highlighting the critical role that autophagy plays in maintaining tubular cell integrity during stress conditions.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Scribble participates in Hippo signaling and is required for normal zebrafish pronephros development

Kassiani Skouloudaki; Michael Puetz; Matias Simons; Jean-Remy Courbard; Christopher Boehlke; Björn Hartleben; Christina Engel; Marcus J. Moeller; Christoph Englert; Frank Bollig; Tobias Schäfer; Marek Mlodzik; Tobias B. Huber; E. Wolfgang Kuehn; Emily Kim; Albrecht Kramer-Zucker; Gerd Walz

Spatial organization of cells and their appendages is controlled by the planar cell polarity pathway, a signaling cascade initiated by the protocadherin Fat in Drosophila. Vertebrates express 4 Fat molecules, Fat1–4. We found that depletion of Fat1 caused cyst formation in the zebrafish pronephros. Knockdown of the PDZ domain containing the adaptor protein Scribble intensified the cyst-promoting phenotype of Fat1 depletion, suggesting that Fat1 and Scribble act in overlapping signaling cascades during zebrafish pronephros development. Supporting the genetic interaction with Fat1, Scribble recognized the PDZ-binding site of Fat1. Depletion of Yes-associated protein 1 (YAP1), a transcriptional co-activator inhibited by Hippo signaling, ameliorated the cyst formation in Fat1-deficient zebrafish, whereas Scribble inhibited the YAP1-induced cyst formation. Thus, reduced Hippo signaling and subsequent YAP1 disinhibition seem to play a role in the development of pronephric cysts after depletion of Fat1 or Scribble. We hypothesize that Hippo signaling is required for normal pronephros development in zebrafish and that Scribble is a candidate link between Fat and the Hippo signaling cascade in vertebrates.


Journal of The American Society of Nephrology | 2014

Unraveling the Role of Podocyte Turnover in Glomerular Aging and Injury

Nicola Wanner; Björn Hartleben; Nadja Herbach; Markus Goedel; Natalie Stickel; Robert Zeiser; Gerd Walz; Marcus J. Moeller; Florian Grahammer; Tobias B. Huber

Podocyte loss is a major determinant of progressive CKD. Although recent studies showed that a subset of parietal epithelial cells can serve as podocyte progenitors, the role of podocyte turnover and regeneration in repair, aging, and nephron loss remains unclear. Here, we combined genetic fate mapping with highly efficient podocyte isolation protocols to precisely quantify podocyte turnover and regeneration. We demonstrate that parietal epithelial cells can give rise to fully differentiated visceral epithelial cells indistinguishable from resident podocytes and that limited podocyte renewal occurs in a diphtheria toxin model of acute podocyte ablation. In contrast, the compensatory programs initiated in response to nephron loss evoke glomerular hypertrophy, but not de novo podocyte generation. In addition, no turnover of podocytes could be detected in aging mice under physiologic conditions. In the absence of podocyte replacement, characteristic features of aging mouse kidneys included progressive accumulation of oxidized proteins, deposits of protein aggregates, loss of podocytes, and glomerulosclerosis. In summary, quantitative investigation of podocyte regeneration in vivo provides novel insights into the mechanism and capacity of podocyte turnover and regeneration in mice. Our data reveal that podocyte generation is mainly confined to glomerular development and may occur after acute glomerular injury, but it fails to regenerate podocytes in aging kidneys or in response to nephron loss.

Collaboration


Dive into the Björn Hartleben's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gerd Walz

University of Freiburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrey S. Shaw

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Miriam Schmidts

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Dontscho Kerjaschki

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Jeffrey H. Miner

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge