Clinical and translational research in acute kidney injury (AKI) requires access to well characterized patients with longitudinal follow up coupled with biological samples enabling investigators to probe the underlying mechanisms and pathways contributing to outcomes.
The specific aims of the Resource for Clinical Studies of AKI (Core A) are to (i) facilitate and support the design and conduct of clinical research in AKI with appropriate tools to collect and record information for detailed phenotypic characterization of patients through the course of AKI, (ii) provide access to comprehensive datasets of well characterized patients with and without AKI, and (iii) provide a biological sample repository that includes human kidney tissue, blood and urine, linked to an accessible clinical database of patients with and without AKI to enable translational research studies. This core will specifically provide investigators access to patients with AKI through an established international network of collaborating investigators who are contributing to an ongoing registry of AKI in the ICU setting currently with over 2,200 patients. Core A is designed to facilitate both hypothesis-driven and hypothesis-generating clinical research by providing essential resources for clinical investigation and enhancing interactions with a large worldwide network of investigators and centers focused on AKI. Since its inception the Core has been very successful in supporting investigators for clinical and translational research. We have established a robust data management system and database of patients with AKI that is flexible in accommodating the research objectives of individual investigators and collaborating centers. A new service being offered by Core A is the addition of human kidney tissues along with biological samples (blood, urine and DNA) from patients with and without AKI and healthy controls. Over the last 4 years, Core A has been highly productive in assisting multiple facets of clinical research supporting 72 investigators including 8 pilot and catalyst awardees and 111 different projects. These combined efforts have resulted in 40 peer-reviewed publications. All services provided by Core A are being utilized by our investigator base. We envision that these rich resources will continue to enable interdisciplinary clinical investigation in AKI to advance our understanding of the natural history and pathophysiology of human AKI. We are confident that Core A in conjunction with the existing resources at UAB and UCSD and other cores within the O'Brien Center will accelerate the translation of new investigative insights towards improving outcomes for patients with AKI.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Center Core Grants (P30)
Project #
5P30DK079337-13
Application #
9972913
Study Section
Special Emphasis Panel (ZDK1)
Project Start
2008-09-01
Project End
2023-07-31
Budget Start
2020-08-01
Budget End
2021-07-31
Support Year
13
Fiscal Year
2020
Total Cost
Indirect Cost
Name
University of Alabama Birmingham
Department
Type
DUNS #
063690705
City
Birmingham
State
AL
Country
United States
Zip Code
35294
Layton, Anita T; Vallon, Volker (2018) SGLT2 inhibition in a kidney with reduced nephron number: modeling and analysis of solute transport and metabolism. Am J Physiol Renal Physiol 314:F969-F984
Neyra, Javier A; Leaf, David E (2018) Risk Prediction Models for Acute Kidney Injury in Critically Ill Patients: Opus in Progressu. Nephron 140:99-104
Patel, Mikita; Yarlagadda, Vidhush; Adedoyin, Oreoluwa et al. (2018) Oxalate induces mitochondrial dysfunction and disrupts redox homeostasis in a human monocyte derived cell line. Redox Biol 15:207-215
Feng, Di; Notbohm, Jacob; Benjamin, Ava et al. (2018) Disease-causing mutation in ?-actinin-4 promotes podocyte detachment through maladaptation to periodic stretch. Proc Natl Acad Sci U S A 115:1517-1522
Rieg, Timo; Vallon, Volker (2018) Development of SGLT1 and SGLT2 inhibitors. Diabetologia 61:2079-2086
Yeboah, Michael M; Hye Khan, Md Abdul; Chesnik, Marla A et al. (2018) Role of the cytochrome P-450/ epoxyeicosatrienoic acids pathway in the pathogenesis of renal dysfunction in cirrhosis. Nephrol Dial Transplant 33:1333-1343
Guan, Z; Wang, F; Cui, X et al. (2018) Mechanisms of sphingosine-1-phosphate-mediated vasoconstriction of rat afferent arterioles. Acta Physiol (Oxf) 222:
Adedoyin, Oreoluwa; Boddu, Ravindra; Traylor, Amie et al. (2018) Heme oxygenase-1 mitigates ferroptosis in renal proximal tubule cells. Am J Physiol Renal Physiol 314:F702-F714
Layton, Anita T; Edwards, Aurélie; Vallon, Volker (2018) Renal potassium handling in rats with subtotal nephrectomy: modeling and analysis. Am J Physiol Renal Physiol 314:F643-F657
Li, Mao; Boddeda, Srinivasa Rao; Chen, Bo et al. (2018) NK cell and Th17 responses are differentially induced in murine cytomegalovirus infected renal allografts and vary according to recipient virus dose and strain. Am J Transplant 18:2647-2662

Showing the most recent 10 out of 404 publications