Abstract

Renal microvascular rarefaction plays a pivotal role in progressive kidney disease. Therefore, modalities to visualize the microcirculation of the kidney will increase our understanding of disease mechanisms and consequently may provide new approaches for evaluating cell-based therapy. At the moment, however, clinical practice is lacking non-invasive, safe, and efficient imaging modalities to monitor renal microvascular changes over time in patients suffering from renal disease. To emphasize the importance, we summarize current knowledge of the renal microcirculation and discussed the involvement in progressive kidney disease. Moreover, an overview of available imaging techniques to uncover renal microvascular morphology, function, and behavior is presented with the associated benefits and limitations. Ultimately, the necessity to assess and investigate renal disease based on in vivo readouts with a resolution up to capillary level may provide a paradigm shift for diagnosis and therapy in the field of nephrology.

Highlights

  • The Renal Blood CirculationThe microvasculature of the human body consists of arterioles, capillaries, and venules and effectuates the exchange of oxygen, nutrients, and metabolites between the blood and the surrounding tissue [8,9]

  • Renal microvascular rarefaction plays a pivotal role in progressive kidney disease

  • It seems that the descending vasa recta (DVR) of the vascular bundles situated in the inner stripe of the outer medulla is responsible for controlling the medullary blood flow (MBF) which may explain the high number of pericytes wrapping this vessel compartment

Read more

Summary

The Renal Blood Circulation

The microvasculature of the human body consists of arterioles, capillaries, and venules and effectuates the exchange of oxygen, nutrients, and metabolites between the blood and the surrounding tissue [8,9]. The main responsibility of arterioles is to regulate the blood flow by adjusting the resistance to ensure that the vital exchange at the level of capillaries can be executed [10] In this regard, continuous adaption to the homeostatic demand of the underlying tissue is mainly dependent on the dynamic plasticity of endothelial cells. Blood drains into the venous system which runs in parallel to the arterial network exiting the kidney via the interlobular, arcuate, interlobar, segmental, and eventually the renal vein right above the ureter. Speaking, this basic vascular pattern is preserved across mammals [14,15]

The Capillary Networks of the Kidney
Endothelial Dysfunction
Pericyte Involvement in Renal Malfunction
Endothelial Cell-Pericyte Signaling Interactions
Pericytes as Precursor of Myofibroblasts
Vascular Imaging Modalities
Ex Vivo
In Vivo
Ultrasound
Findings
Conclusions and Perspectives
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call