Skip to main content
Top
Published in: BMC Nephrology 1/2019

Open Access 01-12-2019 | Polycystic Kidney Disease | Research article

Plasma metabolites and lipids associate with kidney function and kidney volume in hypertensive ADPKD patients early in the disease course

Authors: Kyoungmi Kim, Josephine F. Trott, Guimin Gao, Arlene Chapman, Robert H. Weiss

Published in: BMC Nephrology | Issue 1/2019

Login to get access

Abstract

Background

Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary kidney disease and is characterized by gradual cyst growth and expansion, increase in kidney volume with an ultimate decline in kidney function leading to end stage renal disease (ESRD). Given the decades long period of stable kidney function while cyst growth occurs, it is important to identify those patients who will progress to ESRD. Recent data from our and other laboratories have demonstrated that metabolic reprogramming may play a key role in cystic epithelial proliferation resulting in cyst growth in ADPKD. Height corrected total kidney volume (ht-TKV) accurately reflects cyst burden and predicts future loss of kidney function. We hypothesize that specific plasma metabolites will correlate with eGFR and ht-TKV early in ADPKD, both predictors of disease progression, potentially indicative of early physiologic derangements of renal disease severity.

Methods

To investigate the predictive role of plasma metabolites on eGFR and/or ht-TKV, we used a non-targeted GC-TOF/MS-based metabolomics approach on hypertensive ADPKD patients in the early course of their disease. Patient data was obtained from the HALT-A randomized clinical trial at baseline including estimated glomerular filtration rate (eGFR) and measured ht-TKV. To identify individual metabolites whose intensities are significantly correlated with eGFR and ht-TKV, association analyses were performed using linear regression with each metabolite signal level as the primary predictor variable and baseline eGFR and ht-TKV as the continuous outcomes of interest, while adjusting for covariates. Significance was determined by Storey’s false discovery rate (FDR) q-values to correct for multiple testing.

Results

Twelve metabolites significantly correlated with eGFR and two triglycerides significantly correlated with baseline ht-TKV at FDR q-value < 0.05. Specific significant metabolites, including pseudo-uridine, indole-3-lactate, uric acid, isothreonic acid, and creatinine, have been previously shown to accumulate in plasma and/or urine in both diabetic and cystic renal diseases with advanced renal insufficiency.

Conclusions

This study identifies metabolic derangements in early ADPKD which may be prognostic for ADPKD disease progression.

Clinical trial

HALT Progression of Polycystic Kidney Disease (HALT PKD) Study A; Clinical www.​clinicaltrials.​gov identifier: NCT00283686; first posted January 30, 2006, last update posted March 19, 2015.
Appendix
Available only for authorised users
Literature
1.
go back to reference Consortium TEPKD. The polycystic kidney disease 1 gene encodes a 14 kb transcript and lies within a duplicated region on chromosome 16. Cell. 1994;77:881–94.CrossRef Consortium TEPKD. The polycystic kidney disease 1 gene encodes a 14 kb transcript and lies within a duplicated region on chromosome 16. Cell. 1994;77:881–94.CrossRef
2.
go back to reference Mochizuki T, Wu G, Hayashi T, et al. PKD2, a gene for polycystic kidney disease that encodes an integral membrane protein. Science (New York, NY). 1996;272:1339–42.CrossRef Mochizuki T, Wu G, Hayashi T, et al. PKD2, a gene for polycystic kidney disease that encodes an integral membrane protein. Science (New York, NY). 1996;272:1339–42.CrossRef
3.
go back to reference Porath B, Gainullin VG, Cornec-Le Gall E, et al. Mutations in GANAB, encoding the glucosidase IIalpha subunit, cause autosomal-dominant polycystic kidney and liver disease. Am J Hum Genet. 2016;98:1193–207.CrossRef Porath B, Gainullin VG, Cornec-Le Gall E, et al. Mutations in GANAB, encoding the glucosidase IIalpha subunit, cause autosomal-dominant polycystic kidney and liver disease. Am J Hum Genet. 2016;98:1193–207.CrossRef
4.
go back to reference Qian F, Germino FJ, Cai Y, Zhang X, Somlo S, Germino GG. PKD1 interacts with PKD2 through a probable coiled-coil domain. Nat Genet. 1997;16:179–83.CrossRef Qian F, Germino FJ, Cai Y, Zhang X, Somlo S, Germino GG. PKD1 interacts with PKD2 through a probable coiled-coil domain. Nat Genet. 1997;16:179–83.CrossRef
5.
go back to reference Boletta A, Qian F, Onuchic LF, et al. Polycystin-1, the gene product of PKD1, induces resistance to apoptosis and spontaneous tubulogenesis in MDCK cells. MolCell. 2000;6:1267–73. Boletta A, Qian F, Onuchic LF, et al. Polycystin-1, the gene product of PKD1, induces resistance to apoptosis and spontaneous tubulogenesis in MDCK cells. MolCell. 2000;6:1267–73.
6.
go back to reference Braun WE, Abebe KZ, Brosnahan G, et al. ADPKD progression in patients with no apparent family history and no mutation detected by sanger sequencing. Am J Kidney Dis. 2018;71:294–6.CrossRef Braun WE, Abebe KZ, Brosnahan G, et al. ADPKD progression in patients with no apparent family history and no mutation detected by sanger sequencing. Am J Kidney Dis. 2018;71:294–6.CrossRef
7.
go back to reference Chebib FT, Torres VE. Autosomal dominant polycystic kidney disease: Core curriculum 2016. Am J Kidney Dis. 2016;67:792–810.CrossRef Chebib FT, Torres VE. Autosomal dominant polycystic kidney disease: Core curriculum 2016. Am J Kidney Dis. 2016;67:792–810.CrossRef
8.
go back to reference Ganti S, Taylor S, Kim K, et al. Urinary acylcarnitines are altered in kidney cancer. IntJCancer. 2012;130:2791–800. Ganti S, Taylor S, Kim K, et al. Urinary acylcarnitines are altered in kidney cancer. IntJCancer. 2012;130:2791–800.
9.
go back to reference Chapman AB, Johnson AM, Gabow PA, Schrier RW. Overt proteinuria and microalbuminuria in autosomal dominant polycystic kidney disease. Journal of the American Society of Nephrology : JASN. 1994;5:1349–54.PubMed Chapman AB, Johnson AM, Gabow PA, Schrier RW. Overt proteinuria and microalbuminuria in autosomal dominant polycystic kidney disease. Journal of the American Society of Nephrology : JASN. 1994;5:1349–54.PubMed
10.
go back to reference Torres VE, Grantham JJ, Chapman AB, et al. Potentially modifiable factors affecting the progression of autosomal dominant polycystic kidney disease. Clinical journal of the American Society of Nephrology : CJASN. 2011;6:640–7.CrossRef Torres VE, Grantham JJ, Chapman AB, et al. Potentially modifiable factors affecting the progression of autosomal dominant polycystic kidney disease. Clinical journal of the American Society of Nephrology : CJASN. 2011;6:640–7.CrossRef
11.
go back to reference Gabow PA, Johnson AM, Kaehny WD, et al. Factors affecting the progression of renal disease in autosomal-dominant polycystic kidney disease. Kidney Int. 1992;41:1311–9.CrossRef Gabow PA, Johnson AM, Kaehny WD, et al. Factors affecting the progression of renal disease in autosomal-dominant polycystic kidney disease. Kidney Int. 1992;41:1311–9.CrossRef
12.
go back to reference Schrier RW, Abebe KZ, Perrone RD, et al. Blood pressure in early autosomal dominant polycystic kidney disease. NEnglJMed. 2014;371:2255–66.CrossRef Schrier RW, Abebe KZ, Perrone RD, et al. Blood pressure in early autosomal dominant polycystic kidney disease. NEnglJMed. 2014;371:2255–66.CrossRef
13.
go back to reference Flowers EM, Sudderth J, Zacharias L, et al. Lkb1 deficiency confers glutamine dependency in polycystic kidney disease. Nat Commun. 2018;9:814.CrossRef Flowers EM, Sudderth J, Zacharias L, et al. Lkb1 deficiency confers glutamine dependency in polycystic kidney disease. Nat Commun. 2018;9:814.CrossRef
14.
go back to reference Rowe I, Chiaravalli M, Mannella V, et al. Defective glucose metabolism in polycystic kidney disease identifies a new therapeutic strategy. Nat Med. 2013;19:488–93.CrossRef Rowe I, Chiaravalli M, Mannella V, et al. Defective glucose metabolism in polycystic kidney disease identifies a new therapeutic strategy. Nat Med. 2013;19:488–93.CrossRef
15.
go back to reference Trott JF, Hwang VJ, Ishimaru T, et al. Arginine reprogramming in ADPKD results in arginine-dependent cystogenesis. Am J Physiol Renal Physiol. 2018. Trott JF, Hwang VJ, Ishimaru T, et al. Arginine reprogramming in ADPKD results in arginine-dependent cystogenesis. Am J Physiol Renal Physiol. 2018.
16.
go back to reference Hwang VJ, Kim J, Rand A, et al. The cpk model of recessive PKD shows glutamine dependence associated with the production of the oncometabolite 2-hydroxyglutarate. American Journal of Physiology: Renal Physiology. 2015;309:F492–F8.PubMed Hwang VJ, Kim J, Rand A, et al. The cpk model of recessive PKD shows glutamine dependence associated with the production of the oncometabolite 2-hydroxyglutarate. American Journal of Physiology: Renal Physiology. 2015;309:F492–F8.PubMed
17.
go back to reference Yu ASL, Shen C, Landsittel DP, et al. Baseline total kidney volume and the rate of kidney growth are associated with chronic kidney disease progression in autosomal dominant polycystic kidney disease. Kidney Int. 2018;93:691–9.CrossRef Yu ASL, Shen C, Landsittel DP, et al. Baseline total kidney volume and the rate of kidney growth are associated with chronic kidney disease progression in autosomal dominant polycystic kidney disease. Kidney Int. 2018;93:691–9.CrossRef
18.
go back to reference Perrone RD, Mouksassi MS, Romero K, et al. Total kidney volume is a prognostic biomarker of renal function decline and progression to end-stage renal disease in patients with autosomal dominant polycystic kidney disease. Kidney international reports. 2017;2:442–50.CrossRef Perrone RD, Mouksassi MS, Romero K, et al. Total kidney volume is a prognostic biomarker of renal function decline and progression to end-stage renal disease in patients with autosomal dominant polycystic kidney disease. Kidney international reports. 2017;2:442–50.CrossRef
19.
go back to reference Torres VE, Abebe KZ, Chapman AB, et al. Angiotensin blockade in late autosomal dominant polycystic kidney disease. NEnglJMed. 2014;371:2267–76.CrossRef Torres VE, Abebe KZ, Chapman AB, et al. Angiotensin blockade in late autosomal dominant polycystic kidney disease. NEnglJMed. 2014;371:2267–76.CrossRef
20.
go back to reference Fiehn O. Metabolomics by gas chromatography-mass spectrometry: combined targeted and untargeted profiling. Current protocols in molecular biology 2016;114:30.4.1–.4.2. Fiehn O. Metabolomics by gas chromatography-mass spectrometry: combined targeted and untargeted profiling. Current protocols in molecular biology 2016;114:30.4.1–.4.2.
21.
go back to reference Cajka T, Smilowitz JT, Fiehn O. Validating quantitative untargeted Lipidomics across nine liquid chromatography-high-resolution mass spectrometry platforms. Anal Chem. 2017;89:12360–8.CrossRef Cajka T, Smilowitz JT, Fiehn O. Validating quantitative untargeted Lipidomics across nine liquid chromatography-high-resolution mass spectrometry platforms. Anal Chem. 2017;89:12360–8.CrossRef
22.
go back to reference Johnson JB, Omland KS. Model selection in ecology and evolution. Trends Ecol Evol. 2004;19:101–8.CrossRef Johnson JB, Omland KS. Model selection in ecology and evolution. Trends Ecol Evol. 2004;19:101–8.CrossRef
23.
go back to reference Storey J. A direct approach to false discovery rates. J Royal Stat Soc B. 2002;64:479–98.CrossRef Storey J. A direct approach to false discovery rates. J Royal Stat Soc B. 2002;64:479–98.CrossRef
24.
go back to reference Mao Z, Xie G, Ong AC. Metabolic abnormalities in autosomal dominant polycystic kidney disease. Nephrol Dial Transplant. 2015;30:197–203.CrossRef Mao Z, Xie G, Ong AC. Metabolic abnormalities in autosomal dominant polycystic kidney disease. Nephrol Dial Transplant. 2015;30:197–203.CrossRef
25.
go back to reference Torres VE, Wilson DM, Hattery RR, Segura JW. Renal stone disease in autosomal dominant polycystic kidney disease. Am J Kidney Dis. 1993;22:513–9.CrossRef Torres VE, Wilson DM, Hattery RR, Segura JW. Renal stone disease in autosomal dominant polycystic kidney disease. Am J Kidney Dis. 1993;22:513–9.CrossRef
26.
go back to reference Masoumi A, Reed-Gitomer B, Kelleher C, Bekheirnia MR, Schrier RW. Developments in the management of autosomal dominant polycystic kidney disease. Ther Clin Risk Manag. 2008;4:393–407.CrossRef Masoumi A, Reed-Gitomer B, Kelleher C, Bekheirnia MR, Schrier RW. Developments in the management of autosomal dominant polycystic kidney disease. Ther Clin Risk Manag. 2008;4:393–407.CrossRef
27.
go back to reference Nishiura JL, Neves RF, Eloi SR, Cintra SM, Ajzen SA, Heilberg IP. Evaluation of nephrolithiasis in autosomal dominant polycystic kidney disease patients. Clin J Am Soc Nephrol. 2009;4:838–44.CrossRef Nishiura JL, Neves RF, Eloi SR, Cintra SM, Ajzen SA, Heilberg IP. Evaluation of nephrolithiasis in autosomal dominant polycystic kidney disease patients. Clin J Am Soc Nephrol. 2009;4:838–44.CrossRef
28.
go back to reference Idrizi A, Barbullushi M, Koroshi A, et al. Urinary tract infections in polycystic kidney disease. Med Arh. 2011;65:213–5.CrossRef Idrizi A, Barbullushi M, Koroshi A, et al. Urinary tract infections in polycystic kidney disease. Med Arh. 2011;65:213–5.CrossRef
29.
go back to reference Mejias E, Navas J, Lluberes R, Martinez-Maldonado M. Hyperuricemia, gout, and autosomal dominant polycystic kidney disease. Am J Med Sci. 1989;297:145–8.CrossRef Mejias E, Navas J, Lluberes R, Martinez-Maldonado M. Hyperuricemia, gout, and autosomal dominant polycystic kidney disease. Am J Med Sci. 1989;297:145–8.CrossRef
30.
go back to reference Pavik I, Jaeger P, Kistler AD, et al. Patients with autosomal dominant polycystic kidney disease have elevated fibroblast growth factor 23 levels and a renal leak of phosphate. Kidney Int. 2011;79:234–40.CrossRef Pavik I, Jaeger P, Kistler AD, et al. Patients with autosomal dominant polycystic kidney disease have elevated fibroblast growth factor 23 levels and a renal leak of phosphate. Kidney Int. 2011;79:234–40.CrossRef
31.
go back to reference Allen E, Piontek KB, Garrett-Mayer E, Garcia-Gonzalez M, Gorelick KL, Germino GG. Loss of polycystin-1 or polycystin-2 results in dysregulated apolipoprotein expression in murine tissues via alterations in nuclear hormone receptors. Hum Mol Genet. 2006;15:11–21.CrossRef Allen E, Piontek KB, Garrett-Mayer E, Garcia-Gonzalez M, Gorelick KL, Germino GG. Loss of polycystin-1 or polycystin-2 results in dysregulated apolipoprotein expression in murine tissues via alterations in nuclear hormone receptors. Hum Mol Genet. 2006;15:11–21.CrossRef
32.
go back to reference Menezes LF, Lin CC, Zhou F, Germino GG. Fatty acid oxidation is impaired in an orthologous mouse model of autosomal dominant polycystic kidney disease. EBioMedicine. 2016;5:183–92.CrossRef Menezes LF, Lin CC, Zhou F, Germino GG. Fatty acid oxidation is impaired in an orthologous mouse model of autosomal dominant polycystic kidney disease. EBioMedicine. 2016;5:183–92.CrossRef
33.
go back to reference Chiaravalli M, Rowe I, Mannella V, et al. 2-Deoxy-d-glucose ameliorates PKD progression. J Am Soc Nephrol. 2016;27:1958–69.CrossRef Chiaravalli M, Rowe I, Mannella V, et al. 2-Deoxy-d-glucose ameliorates PKD progression. J Am Soc Nephrol. 2016;27:1958–69.CrossRef
34.
go back to reference Weiss RH, Kim K. Metabolomics in the study of kidney diseases. NatRevNephrology. 2011;8:22–33. Weiss RH, Kim K. Metabolomics in the study of kidney diseases. NatRevNephrology. 2011;8:22–33.
35.
go back to reference Wettersten HI, Weiss RH. Applications of metabolomics for kidney disease research: from biomarkers to therapeutic targets. Organogenesis. 2013;9. Wettersten HI, Weiss RH. Applications of metabolomics for kidney disease research: from biomarkers to therapeutic targets. Organogenesis. 2013;9.
36.
go back to reference Menezes LF, Zhou F, Patterson AD, et al. Network analysis of a Pkd1-mouse model of autosomal dominant polycystic kidney disease identifies HNF4alpha as a disease modifier. PLoS Genet. 2012;8:e1003053.CrossRef Menezes LF, Zhou F, Patterson AD, et al. Network analysis of a Pkd1-mouse model of autosomal dominant polycystic kidney disease identifies HNF4alpha as a disease modifier. PLoS Genet. 2012;8:e1003053.CrossRef
37.
go back to reference Taylor SL, Ganti S, Bukanov NO, et al. A metabolomics approach using juvenile cystic mice to identify urinary biomarkers and altered pathways in polycystic kidney disease. American Journal of Physiology: Renal Physiology. 2010;298:F909–F22.PubMed Taylor SL, Ganti S, Bukanov NO, et al. A metabolomics approach using juvenile cystic mice to identify urinary biomarkers and altered pathways in polycystic kidney disease. American Journal of Physiology: Renal Physiology. 2010;298:F909–F22.PubMed
38.
go back to reference Niwa T, Takeda N, Yoshizumi H. RNA metabolism in uremic patients: accumulation of modified ribonucleosides in uremic serum. Technical note Kidney Int. 1998;53:1801–6.CrossRef Niwa T, Takeda N, Yoshizumi H. RNA metabolism in uremic patients: accumulation of modified ribonucleosides in uremic serum. Technical note Kidney Int. 1998;53:1801–6.CrossRef
39.
go back to reference Niewczas MA, Sirich TL, Mathew AV, et al. Uremic solutes and risk of end-stage renal disease in type 2 diabetes: metabolomic study. Kidney Int. 2014;85:1214–24.CrossRef Niewczas MA, Sirich TL, Mathew AV, et al. Uremic solutes and risk of end-stage renal disease in type 2 diabetes: metabolomic study. Kidney Int. 2014;85:1214–24.CrossRef
40.
go back to reference Helal I, McFann K, Reed B, Yan XD, Schrier RW, Fick-Brosnahan GM. Serum uric acid, kidney volume and progression in autosomal-dominant polycystic kidney disease. Nephrol Dial Transplant. 2013;28:380–5.CrossRef Helal I, McFann K, Reed B, Yan XD, Schrier RW, Fick-Brosnahan GM. Serum uric acid, kidney volume and progression in autosomal-dominant polycystic kidney disease. Nephrol Dial Transplant. 2013;28:380–5.CrossRef
41.
go back to reference Hosoya T, Ichida K, Tabe A. Sakai O. A study of uric acid metabolism and gouty arthritis in patients with polycystic kidney. Nihon Jinzo Gakkai Shi. 1993;35:43–8.PubMed Hosoya T, Ichida K, Tabe A. Sakai O. A study of uric acid metabolism and gouty arthritis in patients with polycystic kidney. Nihon Jinzo Gakkai Shi. 1993;35:43–8.PubMed
42.
go back to reference Lai S, Mastroluca D, Matino S, et al. Early markers of cardiovascular risk in autosomal dominant polycystic kidney disease. Kidney Blood Press Res. 2017;42:1290–302.CrossRef Lai S, Mastroluca D, Matino S, et al. Early markers of cardiovascular risk in autosomal dominant polycystic kidney disease. Kidney Blood Press Res. 2017;42:1290–302.CrossRef
43.
go back to reference Kind T, Tolstikov V, Fiehn O, Weiss RH. A comprehensive urinary metabolomic approach for identifying kidney cancer. AnalBiochem. 2007;363:185–95. Kind T, Tolstikov V, Fiehn O, Weiss RH. A comprehensive urinary metabolomic approach for identifying kidney cancer. AnalBiochem. 2007;363:185–95.
44.
go back to reference Kim K, Taylor SL, Ganti S, Guo L, Osier MV, Weiss RH. Urine metabolomic analysis identifies potential biomarkers and pathogenic pathways in kidney cancer. OMICS. 2011;15:293–303.CrossRef Kim K, Taylor SL, Ganti S, Guo L, Osier MV, Weiss RH. Urine metabolomic analysis identifies potential biomarkers and pathogenic pathways in kidney cancer. OMICS. 2011;15:293–303.CrossRef
45.
go back to reference Wikoff WR, Nagle MA, Kouznetsova VL, Tsigelny IF, Nigam SK. Untargeted metabolomics identifies enterobiome metabolites and putative uremic toxins as substrates of organic anion transporter 1 (Oat1). J ProteomeRes. 2011;10:2842–51.CrossRef Wikoff WR, Nagle MA, Kouznetsova VL, Tsigelny IF, Nigam SK. Untargeted metabolomics identifies enterobiome metabolites and putative uremic toxins as substrates of organic anion transporter 1 (Oat1). J ProteomeRes. 2011;10:2842–51.CrossRef
46.
go back to reference Klawitter J, Klawitter J, McFann K, et al. Bioactive lipid mediators in polycystic kidney disease. J Lipid Res. 2014;55:1139–49.CrossRef Klawitter J, Klawitter J, McFann K, et al. Bioactive lipid mediators in polycystic kidney disease. J Lipid Res. 2014;55:1139–49.CrossRef
47.
go back to reference Pietrzak-Nowacka M, Safranow K, Byra E, Binczak-Kuleta A, Ciechanowicz A, Ciechanowski K. Metabolic syndrome components in patients with autosomal-dominant polycystic kidney disease. Kidney Blood Press Res. 2009;32:405–10.CrossRef Pietrzak-Nowacka M, Safranow K, Byra E, Binczak-Kuleta A, Ciechanowicz A, Ciechanowski K. Metabolic syndrome components in patients with autosomal-dominant polycystic kidney disease. Kidney Blood Press Res. 2009;32:405–10.CrossRef
48.
go back to reference Veeramuthumari P, Isabel W. Clinical study on autosomal dominant polycystic kidney disease among south Indians. International Journal of Clinical Medicine. 2013;2013:200–4.CrossRef Veeramuthumari P, Isabel W. Clinical study on autosomal dominant polycystic kidney disease among south Indians. International Journal of Clinical Medicine. 2013;2013:200–4.CrossRef
49.
go back to reference Barter P. Lipoprotein metabolism and CKD: overview. Clin Exp Nephrol. 2014;18:243–6.CrossRef Barter P. Lipoprotein metabolism and CKD: overview. Clin Exp Nephrol. 2014;18:243–6.CrossRef
50.
go back to reference Hocher B, Adamski J. Metabolomics for clinical use and research in chronic kidney disease. Nat Rev Nephrol. 2017;13:269–84.CrossRef Hocher B, Adamski J. Metabolomics for clinical use and research in chronic kidney disease. Nat Rev Nephrol. 2017;13:269–84.CrossRef
Metadata
Title
Plasma metabolites and lipids associate with kidney function and kidney volume in hypertensive ADPKD patients early in the disease course
Authors
Kyoungmi Kim
Josephine F. Trott
Guimin Gao
Arlene Chapman
Robert H. Weiss
Publication date
01-12-2019
Publisher
BioMed Central
Published in
BMC Nephrology / Issue 1/2019
Electronic ISSN: 1471-2369
DOI
https://doi.org/10.1186/s12882-019-1249-6

Other articles of this Issue 1/2019

BMC Nephrology 1/2019 Go to the issue
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discuss last year's major advances in heart failure and cardiomyopathies.