Advances in Renal Epithelial Cells

A special issue of Cells (ISSN 2073-4409).

Deadline for manuscript submissions: closed (15 September 2023) | Viewed by 3114

Special Issue Editors


E-Mail Website
Guest Editor
Department of Clinical and Experimental Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
Interests: acute kidney injury; chronic kidney injury; kidney cancer; renal progenitors
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Department of Clinical and Experimental Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
Interests: acute kidney injury; podocyte; podocytopathies; renal progenitors
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

The renal epithelial cells that compose the nephron play important roles in physiological processes, from body fluid regulation to ion and acid/base homeostasis, as well as in pathological processes, during kidney repair following injury or tumorigenesis. The advent of new technologies, such as spatial transcriptomics or 3D cultures, have provided researchers with powerful tools to study cell properties and functions, and contributed to the rapid expansion of our knowledge on renal epithelial cells. Recent findings led to a major appreciation of the significance of renal epithelial cell interactions with other cellular components of the kidney, and of the complexity of their response to the microenvironment. Sex-oriented studies have uncovered sex-specific differences in renal epithelial cells, boosting research interest on molecular, cellular and systems-based differences between males and females.

The aim of this Special Issue “Advances in Renal Epithelial Cells” is to assemble the latest studies highlighting the plurality of roles of renal epithelial cells in kidney health and disease. We welcome research papers and reviews, focusing on, but not exclusively, cellular and molecular mechanisms, use of biomaterials, precision medicine, patient-derived organoids and 3D models, animal models, therapeutic strategies and drug testing, single cell/nucleus RNA sequencing, multi-omics.

We hope that this special issue will provide a comprehensive understanding of renal epithelial cells, that will be of interest to both basic and clinician scientists.

Looking forward to receiving your contribution.

Dr. Anna Julie Peired
Dr. Laura Lasagni
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cells is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • epithelial cell
  • kidney injury
  • renal physiology
  • organoids
  • 3D models
  • biomaterials
  • precision medicine
  • molecular pathways
  • sex-based differences

Published Papers (2 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

14 pages, 6281 KiB  
Article
Kidney-Specific Membrane-Bound Serine Proteases CAP1/Prss8 and CAP3/St14 Affect ENaC Subunit Abundances but Not Its Activity
by Elodie Ehret, Sévan Stroh, Muriel Auberson, Frédérique Ino, Yannick Jäger, Marc Maillard, Roman Szabo, Thomas H. Bugge, Simona Frateschi and Edith Hummler
Cells 2023, 12(19), 2342; https://doi.org/10.3390/cells12192342 - 23 Sep 2023
Viewed by 1126
Abstract
The serine proteases CAP1/Prss8 and CAP3/St14 are identified as ENaC channel-activating proteases in vitro, highly suggesting that they are required for proteolytic activation of ENaC in vivo. The present study tested whether CAP3/St14 is relevant for renal proteolytic ENaC activation and affects ENaC-mediated [...] Read more.
The serine proteases CAP1/Prss8 and CAP3/St14 are identified as ENaC channel-activating proteases in vitro, highly suggesting that they are required for proteolytic activation of ENaC in vivo. The present study tested whether CAP3/St14 is relevant for renal proteolytic ENaC activation and affects ENaC-mediated Na+ absorption following Na+ deprivation conditions. CAP3/St14 knockout mice exhibit a significant decrease in CAP1/Prss8 protein expression with altered ENaC subunit and decreased pNCC protein abundances but overall maintain sodium balance. RNAscope-based analyses reveal co-expression of CAP3/St14 and CAP1/Prss8 with alpha ENaC in distal tubules of the cortex from wild-type mice. Double CAP1/Prss8; CAP3/St14-deficiency maintained Na+ and K+ balance on a Na+-deprived diet, restored ENaC subunit protein abundances but showed reduced NCC activity under Na+ deprivation. Overall, our data clearly show that CAP3/St14 is not required for direct proteolytic activation of ENaC but for its protein abundance. Our study reveals a complex regulation of ENaC by these serine proteases on the expression level rather than on its proteolytic activation. Full article
(This article belongs to the Special Issue Advances in Renal Epithelial Cells)
Show Figures

Figure 1

Review

Jump to: Research

20 pages, 864 KiB  
Review
Emerging Immunotherapy Approaches for Advanced Clear Cell Renal Cell Carcinoma
by Lingbin Meng, Katharine A. Collier, Peng Wang, Zihai Li, Paul Monk, Amir Mortazavi, Zhiwei Hu, Daniel Spakowicz, Linghua Zheng and Yuanquan Yang
Cells 2024, 13(1), 34; https://doi.org/10.3390/cells13010034 - 22 Dec 2023
Cited by 1 | Viewed by 1722
Abstract
The most common subtype of renal cell carcinoma is clear cell renal cell carcinoma (ccRCC). While localized ccRCC can be cured with surgery, metastatic disease has a poor prognosis. Recently, immunotherapy has emerged as a promising approach for advanced ccRCC. This review provides [...] Read more.
The most common subtype of renal cell carcinoma is clear cell renal cell carcinoma (ccRCC). While localized ccRCC can be cured with surgery, metastatic disease has a poor prognosis. Recently, immunotherapy has emerged as a promising approach for advanced ccRCC. This review provides a comprehensive overview of the evolving immunotherapeutic landscape for metastatic ccRCC. Immune checkpoint inhibitors (ICIs) like PD-1/PD-L1 and CTLA-4 inhibitors have demonstrated clinical efficacy as monotherapies and in combination regimens. Combination immunotherapies pairing ICIs with antiangiogenic agents, other immunomodulators, or novel therapeutic platforms such as bispecific antibodies and chimeric antigen receptor (CAR) T-cell therapy are areas of active research. Beyond the checkpoint blockade, additional modalities including therapeutic vaccines, cytokines, and oncolytic viruses are also being explored for ccRCC. This review discusses the mechanisms, major clinical trials, challenges, and future directions for these emerging immunotherapies. While current strategies have shown promise in improving patient outcomes, continued research is critical for expanding and optimizing immunotherapy approaches for advanced ccRCC. Realizing the full potential of immunotherapy will require elucidating mechanisms of response and resistance, developing predictive biomarkers, and rationally designing combination therapeutic regimens tailored to individual patients. Advances in immunotherapy carry immense promise for transforming the management of metastatic ccRCC. Full article
(This article belongs to the Special Issue Advances in Renal Epithelial Cells)
Show Figures

Figure 1

Back to TopTop