Skip to main content
Top
Published in: Gut Pathogens 1/2016

Open Access 01-12-2016 | Research

Examination of the effects of Campylobacter concisus zonula occludens toxin on intestinal epithelial cells and macrophages

Authors: Vikneswari Mahendran, Fang Liu, Stephen M. Riordan, Michael C. Grimm, Mark M. Tanaka, Li Zhang

Published in: Gut Pathogens | Issue 1/2016

Login to get access

Abstract

Background

Campylobacter concisus is a Gram-negative bacterium that is associated with inflammatory bowel disease (IBD). Some C. concisus strains carry zonula occludens toxin (zot) gene which has polymorphisms. This study investigated the effects of C. concisus Zot on intestinal epithelial cells and macrophages using cell line models.

Methods

Campylobacter concisus zot 808T gene, a polymorphism that is associated with active IBD, was cloned and expressed in Escherichia coli. The effects of C. concisus Zot on intestinal epithelial barrier were examined using Caco-2 cell model. Apoptosis induced by C. concisus Zot in Caco-2 cells was assessed by measuring the levels of caspase 3/7. The production of pro-inflammatory cytokines induced by C. concisus Zot in HT-29 cells and in THP-1 macrophage-like cells was measured using ELISA kits. Whether exposure to C. concisus Zot can affect the responses of macrophages to E. coli K12 was also investigated.

Results

Campylobacter concisus Zot caused prolonged intestinal epithelial barrier damage, induced intestinal epithelial cell apoptosis, induced epithelial production of TNF-α and IL-8 and upregulated TNF-α in THP-1 macrophage-like cells. Pre-exposure to C. concisus Zot significantly enhanced the production of TNF-α and IL-8 as well as phagocytosis by THP-1 macrophage-like cells in response to E. coli K12.

Conclusion

This study suggests that C. concisus Zot may have enteric pathogenic potential by damaging intestinal epithelial barrier, inducing intestinal epithelial and macrophage production of proinflammatory cytokines in particular TNF-α and enhancing the responses of macrophages to other enteric bacterial species.
Appendix
Available only for authorised users
Literature
1.
go back to reference Zhang L, Man SM, Day AS, Leach ST, Lemberg DA, Dutt S, Stormon M, Otley A, O’Loughlin EV, Magoffin A, et al. Detection and isolation of Campylobacter species other than C. jejuni from children with Crohn’s disease. J Clin Microbiol. 2009;47:453–5.CrossRefPubMedPubMedCentral Zhang L, Man SM, Day AS, Leach ST, Lemberg DA, Dutt S, Stormon M, Otley A, O’Loughlin EV, Magoffin A, et al. Detection and isolation of Campylobacter species other than C. jejuni from children with Crohn’s disease. J Clin Microbiol. 2009;47:453–5.CrossRefPubMedPubMedCentral
2.
go back to reference Mahendran V, Riordan SM, Grimm MC, Tran TA, Major J, Kaakoush NO, Mitchell H, Zhang L. Prevalence of Campylobacter species in adult Crohn’s disease and the preferential colonization sites of Campylobacter species in the human intestine. PLoS One. 2011;6:e25417.CrossRefPubMedPubMedCentral Mahendran V, Riordan SM, Grimm MC, Tran TA, Major J, Kaakoush NO, Mitchell H, Zhang L. Prevalence of Campylobacter species in adult Crohn’s disease and the preferential colonization sites of Campylobacter species in the human intestine. PLoS One. 2011;6:e25417.CrossRefPubMedPubMedCentral
3.
go back to reference Mukhopadhya I, Thomson JM, Hansen R, Berry SH, El-Omar EM, Hold GL. Detection of Campylobacter concisus and other Campylobacter species in colonic biopsies from adults with ulcerative colitis. PLoS One. 2011;6:e21490.CrossRefPubMedPubMedCentral Mukhopadhya I, Thomson JM, Hansen R, Berry SH, El-Omar EM, Hold GL. Detection of Campylobacter concisus and other Campylobacter species in colonic biopsies from adults with ulcerative colitis. PLoS One. 2011;6:e21490.CrossRefPubMedPubMedCentral
4.
go back to reference Man SM, Zhang L, Day AS, Leach ST, Lemberg DA, Mitchell H. Campylobacter concisus and other Campylobacter species in children with newly diagnosed Crohn’s disease. Inflamm Bowel Dis. 2010;16:1008–16.CrossRefPubMed Man SM, Zhang L, Day AS, Leach ST, Lemberg DA, Mitchell H. Campylobacter concisus and other Campylobacter species in children with newly diagnosed Crohn’s disease. Inflamm Bowel Dis. 2010;16:1008–16.CrossRefPubMed
5.
go back to reference Zhang L, Budiman V, Day AS, Mitchell H, Lemberg DA, Riordan SM, Grimm M, Leach ST, Ismail Y. Isolation and detection of Campylobacter concisus from saliva of healthy individuals and patients with inflammatory bowel disease. J Clin Microbiol. 2010;48:2965–7.CrossRefPubMedPubMedCentral Zhang L, Budiman V, Day AS, Mitchell H, Lemberg DA, Riordan SM, Grimm M, Leach ST, Ismail Y. Isolation and detection of Campylobacter concisus from saliva of healthy individuals and patients with inflammatory bowel disease. J Clin Microbiol. 2010;48:2965–7.CrossRefPubMedPubMedCentral
6.
go back to reference Tanner ACR, Badger S, Lai C, Listgarten MA, Visconti RA, Socransky SS. Wolinella gen. nov., Wolinella succinogenes (Vibrio succinogenes Wolin et al.) comb. nov., and Description of Bacteroides gracilis sp. nov., Wolinella recta sp. nov., Campylobacter concisus sp. nov., and Eikenella corrodens from humans with periodontal disease. Int J Syst Bacteriol. 1981;31:432–45.CrossRef Tanner ACR, Badger S, Lai C, Listgarten MA, Visconti RA, Socransky SS. Wolinella gen. nov., Wolinella succinogenes (Vibrio succinogenes Wolin et al.) comb. nov., and Description of Bacteroides gracilis sp. nov., Wolinella recta sp. nov., Campylobacter concisus sp. nov., and Eikenella corrodens from humans with periodontal disease. Int J Syst Bacteriol. 1981;31:432–45.CrossRef
7.
go back to reference Ismail Y, Mahendran V, Octavia S, Day AS, Riordan SM, Grimm MC, Lan R, Lemberg D, Tran TA, Zhang L. Investigation of the enteric pathogenic potential of oral Campylobacter concisus strains isolated from patients with inflammatory bowel disease. PLoS One. 2012;7:e38217.CrossRefPubMedPubMedCentral Ismail Y, Mahendran V, Octavia S, Day AS, Riordan SM, Grimm MC, Lan R, Lemberg D, Tran TA, Zhang L. Investigation of the enteric pathogenic potential of oral Campylobacter concisus strains isolated from patients with inflammatory bowel disease. PLoS One. 2012;7:e38217.CrossRefPubMedPubMedCentral
8.
go back to reference Mahendran V, Octavia S, Demirbas OF, Sabrina S, Ma R, Lan R, Riordan SM, Grimm MC, Zhang L. Delineation of genetic relatedness and population structure of oral and enteric Campylobacter concisus strains by analysis of housekeeping genes. Microbiology. 2015;161:1600–12.CrossRefPubMed Mahendran V, Octavia S, Demirbas OF, Sabrina S, Ma R, Lan R, Riordan SM, Grimm MC, Zhang L. Delineation of genetic relatedness and population structure of oral and enteric Campylobacter concisus strains by analysis of housekeeping genes. Microbiology. 2015;161:1600–12.CrossRefPubMed
9.
go back to reference Nielsen HL, Nielsen H, Ejlertsen T, Engberg J, Günzel D, Zeitz M, Hering NA, Fromm M, Schulzke J-D, Bücker R. Oral and fecal Campylobacter concisus strains perturb barrier function by apoptosis induction in HT-29/B6 intestinal epithelial cells. PLoS One. 2011;6:e23858.CrossRefPubMedPubMedCentral Nielsen HL, Nielsen H, Ejlertsen T, Engberg J, Günzel D, Zeitz M, Hering NA, Fromm M, Schulzke J-D, Bücker R. Oral and fecal Campylobacter concisus strains perturb barrier function by apoptosis induction in HT-29/B6 intestinal epithelial cells. PLoS One. 2011;6:e23858.CrossRefPubMedPubMedCentral
11.
go back to reference Lindblom GB, Sjogren E, Hansson-Westerberg J, Kaijser B. Campylobacter upsaliensis, C. sputorum sputorum and C. concisus as common causes of diarrhoea in Swedish children. Scand J Infect Dis. 1995;27:187–8.CrossRefPubMed Lindblom GB, Sjogren E, Hansson-Westerberg J, Kaijser B. Campylobacter upsaliensis, C. sputorum sputorum and C. concisus as common causes of diarrhoea in Swedish children. Scand J Infect Dis. 1995;27:187–8.CrossRefPubMed
12.
go back to reference Nielsen HL, Ejlertsen T, Engberg J, Nielsen H. High incidence of Campylobacter concisus in gastroenteritis in North Jutland, Denmark: a population-based study. Clin Microbiol Infect. 2013;19:445–50.CrossRefPubMed Nielsen HL, Ejlertsen T, Engberg J, Nielsen H. High incidence of Campylobacter concisus in gastroenteritis in North Jutland, Denmark: a population-based study. Clin Microbiol Infect. 2013;19:445–50.CrossRefPubMed
13.
go back to reference Kalischuk L, Inglis GD. Comparative genotypic and pathogenic examination of Campylobacter concisus isolates from diarrheic and non-diarrheic humans. BMC Microbiol. 2011;11:53.CrossRefPubMedPubMedCentral Kalischuk L, Inglis GD. Comparative genotypic and pathogenic examination of Campylobacter concisus isolates from diarrheic and non-diarrheic humans. BMC Microbiol. 2011;11:53.CrossRefPubMedPubMedCentral
14.
go back to reference Lastovica AJ. Emerging Campylobacter spp.: the tip of the iceberg. Clin Microbiol Newsletter. 2006;28:49–56.CrossRef Lastovica AJ. Emerging Campylobacter spp.: the tip of the iceberg. Clin Microbiol Newsletter. 2006;28:49–56.CrossRef
15.
go back to reference Istivan TS, Smith SC, Fry BN, Coloe PJ. Characterization of Campylobacter concisus hemolysins. FEMS Immunol Med Microbiol. 2008;54:224–35.CrossRefPubMed Istivan TS, Smith SC, Fry BN, Coloe PJ. Characterization of Campylobacter concisus hemolysins. FEMS Immunol Med Microbiol. 2008;54:224–35.CrossRefPubMed
16.
go back to reference Kaakoush NO, Man SM, Lamb S, Raftery MJ, Wilkins MR, Kovach Z, Mitchell H. The secretome of Campylobacter concisus. FEBS J. 2010;277:1606–17.CrossRefPubMed Kaakoush NO, Man SM, Lamb S, Raftery MJ, Wilkins MR, Kovach Z, Mitchell H. The secretome of Campylobacter concisus. FEBS J. 2010;277:1606–17.CrossRefPubMed
17.
go back to reference Mahendran V, Tan YS, Riordan SM, Grimm MC, Day AS, Lemberg DA, Octavia S, Lan R, Zhang L. The prevalence and polymorphisms of zonula occluden toxin gene in multiple Campylobacter concisus strains isolated from saliva of patients with inflammatory bowel disease and controls. PLoS One. 2013;8:e75525.CrossRefPubMedPubMedCentral Mahendran V, Tan YS, Riordan SM, Grimm MC, Day AS, Lemberg DA, Octavia S, Lan R, Zhang L. The prevalence and polymorphisms of zonula occluden toxin gene in multiple Campylobacter concisus strains isolated from saliva of patients with inflammatory bowel disease and controls. PLoS One. 2013;8:e75525.CrossRefPubMedPubMedCentral
18.
go back to reference Fasano A, Baudry B, Pumplin DW, Wasserman SS, Tall BD, Ketley JM, Kaper JB. Vibrio cholerae produces a second enterotoxin, which affects intestinal tight junctions. Proc Natl Acad Sci USA. 1991;88:5242–6.CrossRefPubMedPubMedCentral Fasano A, Baudry B, Pumplin DW, Wasserman SS, Tall BD, Ketley JM, Kaper JB. Vibrio cholerae produces a second enterotoxin, which affects intestinal tight junctions. Proc Natl Acad Sci USA. 1991;88:5242–6.CrossRefPubMedPubMedCentral
19.
go back to reference Fasano A, Uzzau S, Fiore C, Margaretten K. The enterotoxic effect of zonula occludens toxin on rabbit small intestine involves the paracellular pathway. Gastroenterology. 1997;112:839–46.CrossRefPubMed Fasano A, Uzzau S, Fiore C, Margaretten K. The enterotoxic effect of zonula occludens toxin on rabbit small intestine involves the paracellular pathway. Gastroenterology. 1997;112:839–46.CrossRefPubMed
20.
go back to reference Zhang L, Lee H, Grimm MC, Riordan SM, Day AS, Lemberg DA. Campylobacter concisus and inflammatory bowel disease. World J Gastroenterol. 2014;20:1259–67.CrossRefPubMedPubMedCentral Zhang L, Lee H, Grimm MC, Riordan SM, Day AS, Lemberg DA. Campylobacter concisus and inflammatory bowel disease. World J Gastroenterol. 2014;20:1259–67.CrossRefPubMedPubMedCentral
21.
go back to reference Ismail Y, Lee H, Riordan SM, Grimm MC, Zhang L. The effects of oral and enteric Campylobacter concisus strains on expression of TLR4, MD-2, TLR2, TLR5 and COX-2 in HT-29 cells. PLoS One. 2013;8:e56888.CrossRefPubMedPubMedCentral Ismail Y, Lee H, Riordan SM, Grimm MC, Zhang L. The effects of oral and enteric Campylobacter concisus strains on expression of TLR4, MD-2, TLR2, TLR5 and COX-2 in HT-29 cells. PLoS One. 2013;8:e56888.CrossRefPubMedPubMedCentral
22.
go back to reference Sambuy Y, Angelis I, Ranaldi G, Scarino ML, Stammati A, Zucco F. The Caco-2 cell line as a model of the intestinal barrier: influence of cell and culture-related factors on Caco-2 cell functional characteristics. Cell Biol Toxicol. 2005;21:1–26.CrossRefPubMed Sambuy Y, Angelis I, Ranaldi G, Scarino ML, Stammati A, Zucco F. The Caco-2 cell line as a model of the intestinal barrier: influence of cell and culture-related factors on Caco-2 cell functional characteristics. Cell Biol Toxicol. 2005;21:1–26.CrossRefPubMed
23.
go back to reference Kwak YK, Vikstrom E, Magnusson KE, Vecsey-Semjen B, Colque-Navarro P, Mollby R. The Staphylococcus aureus alpha-toxin perturbs the barrier function in Caco-2 epithelial cell monolayers by altering junctional integrity. Infect Immun. 2012;80:1670–80.CrossRefPubMedPubMedCentral Kwak YK, Vikstrom E, Magnusson KE, Vecsey-Semjen B, Colque-Navarro P, Mollby R. The Staphylococcus aureus alpha-toxin perturbs the barrier function in Caco-2 epithelial cell monolayers by altering junctional integrity. Infect Immun. 2012;80:1670–80.CrossRefPubMedPubMedCentral
24.
go back to reference Peng L, He Z, Chen W, Holzman IR, Lin J. Effects of butyrate on intestinal barrier function in a Caco-2 cell monolayer model of intestinal barrier. Pediatr Res. 2007;61:37–41.CrossRefPubMed Peng L, He Z, Chen W, Holzman IR, Lin J. Effects of butyrate on intestinal barrier function in a Caco-2 cell monolayer model of intestinal barrier. Pediatr Res. 2007;61:37–41.CrossRefPubMed
25.
go back to reference Man SM, Kaakoush NO, Leach ST, Nahidi L, Lu HK, Norman J, Day AS, Zhang L, Mitchell HM. Host attachment, invasion, and stimulation of proinflammatory cytokines by Campylobacter concisus and other non-Campylobacter jejuni Campylobacter species. J Infect Dis. 2010;202:1855–65.CrossRefPubMed Man SM, Kaakoush NO, Leach ST, Nahidi L, Lu HK, Norman J, Day AS, Zhang L, Mitchell HM. Host attachment, invasion, and stimulation of proinflammatory cytokines by Campylobacter concisus and other non-Campylobacter jejuni Campylobacter species. J Infect Dis. 2010;202:1855–65.CrossRefPubMed
26.
go back to reference Gopalakrishnan S, Pandey N, Tamiz AP, Vere J, Carrasco R, Somerville R, Tripathi A, Ginski M, Paterson BM, Alkan SS. Mechanism of action of ZOT-derived peptide AT-1002, a tight junction regulator and absorption enhancer. Int J Pharm. 2009;365:121–30.CrossRefPubMed Gopalakrishnan S, Pandey N, Tamiz AP, Vere J, Carrasco R, Somerville R, Tripathi A, Ginski M, Paterson BM, Alkan SS. Mechanism of action of ZOT-derived peptide AT-1002, a tight junction regulator and absorption enhancer. Int J Pharm. 2009;365:121–30.CrossRefPubMed
27.
go back to reference Sambuy Y, De Angelis I, Ranaldi G, Scarino ML, Stammati A, Zucco F. The Caco-2 cell line as a model of the intestinal barrier: influence of cell and culture-related factors on Caco-2 cell functional characteristics. Cell Biol Toxicol. 2005;21:1–26.CrossRefPubMed Sambuy Y, De Angelis I, Ranaldi G, Scarino ML, Stammati A, Zucco F. The Caco-2 cell line as a model of the intestinal barrier: influence of cell and culture-related factors on Caco-2 cell functional characteristics. Cell Biol Toxicol. 2005;21:1–26.CrossRefPubMed
28.
go back to reference Hanslick JL, Lau K, Noguchi KK, Olney JW, Zorumski CF, Mennerick S, Farber NB. Dimethyl sulfoxide (DMSO) produces widespread apoptosis in the developing central nervous system. Neurobiol Dis. 2009;34:1–10.CrossRefPubMedPubMedCentral Hanslick JL, Lau K, Noguchi KK, Olney JW, Zorumski CF, Mennerick S, Farber NB. Dimethyl sulfoxide (DMSO) produces widespread apoptosis in the developing central nervous system. Neurobiol Dis. 2009;34:1–10.CrossRefPubMedPubMedCentral
30.
go back to reference Bahrami B, Macfarlane S, Macfarlane GT. Induction of cytokine formation by human intestinal bacteria in gut epithelial cell lines. J Appl Microbiol. 2011;110:353–63.CrossRefPubMed Bahrami B, Macfarlane S, Macfarlane GT. Induction of cytokine formation by human intestinal bacteria in gut epithelial cell lines. J Appl Microbiol. 2011;110:353–63.CrossRefPubMed
31.
go back to reference Eckmann L, Jung HC, Schurer-Maly C, Panja A, Morzycka-Wroblewska E, Kagnoff MF. Differential cytokine expression by human intestinal epithelial cell lines: regulated expression of interleukin 8. Gastroenterology. 1993;105:1689–97.PubMed Eckmann L, Jung HC, Schurer-Maly C, Panja A, Morzycka-Wroblewska E, Kagnoff MF. Differential cytokine expression by human intestinal epithelial cell lines: regulated expression of interleukin 8. Gastroenterology. 1993;105:1689–97.PubMed
32.
go back to reference Ohkusa T, Yoshida T, Sato N, Watanabe S, Tajiri H, Okayasu I. Commensal bacteria can enter colonic epithelial cells and induce proinflammatory cytokine secretion: a possible pathogenic mechanism of ulcerative colitis. J Med Microbiol. 2009;58:535–45.CrossRefPubMedPubMedCentral Ohkusa T, Yoshida T, Sato N, Watanabe S, Tajiri H, Okayasu I. Commensal bacteria can enter colonic epithelial cells and induce proinflammatory cytokine secretion: a possible pathogenic mechanism of ulcerative colitis. J Med Microbiol. 2009;58:535–45.CrossRefPubMedPubMedCentral
33.
go back to reference Daigneault M, Preston JA, Marriott HM, Whyte MK, Dockrell DH. The identification of markers of macrophage differentiation in PMA-stimulated THP-1 cells and monocyte-derived macrophages. PLoS One. 2010;5:e8668.CrossRefPubMedPubMedCentral Daigneault M, Preston JA, Marriott HM, Whyte MK, Dockrell DH. The identification of markers of macrophage differentiation in PMA-stimulated THP-1 cells and monocyte-derived macrophages. PLoS One. 2010;5:e8668.CrossRefPubMedPubMedCentral
34.
go back to reference Harrison LM, van den Hoogen C, van Haaften WC, Tesh VL. Chemokine expression in the monocytic cell line THP-1 in response to purified shiga toxin 1 and/or lipopolysaccharides. Infect Immun. 2005;73:403–12.CrossRefPubMedPubMedCentral Harrison LM, van den Hoogen C, van Haaften WC, Tesh VL. Chemokine expression in the monocytic cell line THP-1 in response to purified shiga toxin 1 and/or lipopolysaccharides. Infect Immun. 2005;73:403–12.CrossRefPubMedPubMedCentral
35.
36.
go back to reference Braegger CP, Nicholls S, Murch SH, Stephens S, MacDonald TT. Tumour necrosis factor alpha in stool as a marker of intestinal inflammation. Lancet. 1992;339:89–91.CrossRefPubMed Braegger CP, Nicholls S, Murch SH, Stephens S, MacDonald TT. Tumour necrosis factor alpha in stool as a marker of intestinal inflammation. Lancet. 1992;339:89–91.CrossRefPubMed
37.
go back to reference Breese EJ, Michie CA, Nicholls SW, Murch SH, Williams CB, Domizio P, Walker-Smith JA, MacDonald TT. Tumor necrosis factor alpha-producing cells in the intestinal mucosa of children with inflammatory bowel disease. Gastroenterology. 1994;106:1455–66.PubMed Breese EJ, Michie CA, Nicholls SW, Murch SH, Williams CB, Domizio P, Walker-Smith JA, MacDonald TT. Tumor necrosis factor alpha-producing cells in the intestinal mucosa of children with inflammatory bowel disease. Gastroenterology. 1994;106:1455–66.PubMed
38.
go back to reference Komatsu M, Kobayashi D, Saito K, Furuya D, Yagihashi A, Araake H, Tsuji N, Sakamaki S, Niitsu Y, Watanabe N. Tumor necrosis factor-α in serum of patients with inflammatory bowel disease as measured by a highly sensitive immuno-PCR. Clin Chem. 2001;47:1297–301.PubMed Komatsu M, Kobayashi D, Saito K, Furuya D, Yagihashi A, Araake H, Tsuji N, Sakamaki S, Niitsu Y, Watanabe N. Tumor necrosis factor-α in serum of patients with inflammatory bowel disease as measured by a highly sensitive immuno-PCR. Clin Chem. 2001;47:1297–301.PubMed
39.
go back to reference Roulis M, Armaka M, Manoloukos M, Apostolaki M, Kollias G, Cominelli F. Intestinal epithelial cells as producers but not targets of chronic TNF suffice to cause murine Crohn-like pathology. Pro Natl Acad Sci USA. 2011;108:5396–401.CrossRef Roulis M, Armaka M, Manoloukos M, Apostolaki M, Kollias G, Cominelli F. Intestinal epithelial cells as producers but not targets of chronic TNF suffice to cause murine Crohn-like pathology. Pro Natl Acad Sci USA. 2011;108:5396–401.CrossRef
40.
go back to reference Hess DJ, Henry-Stanley MJ, Bendel CM, Zhang B, Johnson MA, Wells CL. Escherichia coli and TNF-alpha modulate macrophage phagocytosis of Candida glabrata. J Surg Res. 2009;155:217–24.CrossRefPubMedPubMedCentral Hess DJ, Henry-Stanley MJ, Bendel CM, Zhang B, Johnson MA, Wells CL. Escherichia coli and TNF-alpha modulate macrophage phagocytosis of Candida glabrata. J Surg Res. 2009;155:217–24.CrossRefPubMedPubMedCentral
Metadata
Title
Examination of the effects of Campylobacter concisus zonula occludens toxin on intestinal epithelial cells and macrophages
Authors
Vikneswari Mahendran
Fang Liu
Stephen M. Riordan
Michael C. Grimm
Mark M. Tanaka
Li Zhang
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Gut Pathogens / Issue 1/2016
Electronic ISSN: 1757-4749
DOI
https://doi.org/10.1186/s13099-016-0101-9

Other articles of this Issue 1/2016

Gut Pathogens 1/2016 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 discusses last year's major advances in heart failure and cardiomyopathies.