Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 169(3): 510-522.e20, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28431249

ABSTRACT

Organ-specific functions of tissue-resident macrophages in the steady-state heart are unknown. Here, we show that cardiac macrophages facilitate electrical conduction through the distal atrioventricular node, where conducting cells densely intersperse with elongated macrophages expressing connexin 43. When coupled to spontaneously beating cardiomyocytes via connexin-43-containing gap junctions, cardiac macrophages have a negative resting membrane potential and depolarize in synchrony with cardiomyocytes. Conversely, macrophages render the resting membrane potential of cardiomyocytes more positive and, according to computational modeling, accelerate their repolarization. Photostimulation of channelrhodopsin-2-expressing macrophages improves atrioventricular conduction, whereas conditional deletion of connexin 43 in macrophages and congenital lack of macrophages delay atrioventricular conduction. In the Cd11bDTR mouse, macrophage ablation induces progressive atrioventricular block. These observations implicate macrophages in normal and aberrant cardiac conduction.


Subject(s)
Heart Conduction System , Macrophages/physiology , Animals , Connexin 43/metabolism , Female , Heart Atria/cytology , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myocytes, Cardiac/physiology
2.
J Infect Dis ; 229(5): 1372-1381, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38109685

ABSTRACT

BACKGROUND: Altered mediators of airway tissue remodeling such as matrix metalloproteinases (MMPs) in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection may contribute to morbidity in coronavirus disease 2019 (COVID-19); however, the differential impact of SARS-CoV-2 variants of concern (VOCs) on MMPs is unknown. METHODS: Using both in vitro human airway cell culture model and in vivo transgenic mouse model of SARS-CoV-2 infection, we studied the differential effect of SARS-CoV-2 VOCs on expression of key MMPs and inflammatory mediators in airway cells and tissues. RESULTS: The most consistent findings with all SARS-CoV-2 variants in infected compared to uninfected human bronchial epithelial cell air-liquid interface cultures were the SARS-CoV-2-induced increases in MMP-12 and tissue inhibitor of MMPs. Infection with both SARS-CoV-2 wild type and SARS-CoV-2 Delta variant over 3 days postinfection (dpi) and with Beta variant over 7 dpi increased lung tissue levels of MMP-9 compared to uninfected mice. Overall, SARS-CoV-2 variants had differential dose-dependent impact on secretion of MMP-1, MMP-2, MMP-9, and MMP-12 that varied at the protein versus the gene level and in the early noninflammatory compared to late inflammatory phase of infection. CONCLUSIONS: We provide novel mechanistic insight that the differential impact of SARS-CoV-2 variants on severity of COVID-19 may partially be attributed to unique changes in MMPs.


Subject(s)
COVID-19 , Lung , Matrix Metalloproteinase 12 , Mice, Transgenic , SARS-CoV-2 , Animals , COVID-19/virology , COVID-19/pathology , COVID-19/metabolism , Humans , Mice , Lung/virology , Lung/pathology , Matrix Metalloproteinase 12/metabolism , Matrix Metalloproteinase 12/genetics , Disease Models, Animal , Airway Remodeling , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases/genetics , Epithelial Cells/virology
3.
Am J Transplant ; 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38219867

ABSTRACT

Cardiac allograft vasculopathy (CAV) causes late graft failure and mortality after heart transplantation. Donor-specific antibodies (DSAs) lead to chronic endothelial cell injury, inflammation, and arterial intimal thickening. In this study, GeoMx digital spatial profiling was used to analyze arterial areas of interest (AOIs) from CAV+DSA+ rejected cardiac allografts (N = 3; 22 AOIs total). AOIs were categorized based on CAV neointimal thickening and underwent whole transcriptome and protein profiling. By comparing our transcriptomic data with that of healthy control vessels of rapid autopsy myocardial tissue, we pinpointed specific pathways and transcripts indicative of heightened inflammatory profiles in CAV lesions. Moreover, we identified protein and transcriptomic signatures distinguishing CAV lesions exhibiting low and high neointimal lesions. AOIs with low neointima showed increased markers for activated inflammatory infiltrates, endothelial cell activation transcripts, and gene modules involved in metalloproteinase activation and TP53 regulation of caspases. Inflammatory and apoptotic proteins correlated with inflammatory modules in low neointima AOIs. High neointima AOIs exhibited elevated TGFß-regulated transcripts and modules enriched for platelet activation/aggregation. Proteins associated with growth factors/survival correlated with modules enriched for proliferation/repair in high neointima AOIs. Our findings reveal novel insight into immunological mechanisms mediating CAV pathogenesis.

4.
Am J Transplant ; 24(3): 406-418, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38379280

ABSTRACT

HLA donor-specific antibodies (DSA) elicit alloimmune responses against the graft vasculature, leading to endothelial cell (EC) activation and monocyte infiltration during antibody-mediated rejection (AMR). AMR promotes chronic inflammation and remodeling, leading to thickening of the arterial intima termed transplant vasculopathy or cardiac allograft vasculopathy (CAV) in heart transplants. Intragraft-recipient macrophages serve as a diagnostic marker in AMR; however, their polarization and function remain unclear. In this study, we utilized an in vitro Transwell coculture system to explore the mechanisms of monocyte-to-macrophage polarization induced by HLA I DSA-activated ECs. Anti-HLA I (IgG or F(ab')2) antibody-activated ECs induced the polarization of M2 macrophages with increased CD206 expression and MMP9 secretion. However, inhibition of TLR4 signaling or PSGL-1-P-selectin interactions significantly decreased both CD206 and MMP9. Monocyte adherence to Fc-P-selectin coated plates induced M2 macrophages with increased CD206 and MMP9. Moreover, Fc-receptor and IgG interactions synergistically enhanced active-MMP9 in conjunction with P-selectin. Transcriptomic analysis of arteries from DSA+CAV+ rejected cardiac allografts and multiplex-immunofluorescent staining illustrated the expression of CD68+CD206+CD163+MMP9+ M2 macrophages within the neointima of CAV-affected lesions. These findings reveal a novel mechanism linking HLA I antibody-activated endothelium to the generation of M2 macrophages which secrete vascular remodeling proteins contributing to AMR and CAV pathogenesis.


Subject(s)
Toll-Like Receptor 4 , Vascular Diseases , Humans , Matrix Metalloproteinase 9 , P-Selectin , Macrophages , Endothelium , HLA Antigens , Allografts , Immunoglobulin G
5.
Cell Tissue Res ; 391(1): 189-203, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36376769

ABSTRACT

Autosomal dominant polycystic kidney disease (PKD) is a hereditary kidney disorder which can affect cardiovascular system. Cardiac hypertrophy and cardiomyopathy in PKD have been reported by echocardiography analyses, but histopathology analyses of human PKD hearts have never been examined. The current studies evaluated human heart tissues from five subjects without PKD (non-PKD) and five subjects with PKD. Our histopathology data of human PKD hearts showed an increased extracellular matrix associated with cardiac hypertrophy and fibrosis. Hypertrophy- and fibrosis-associated pathways involving abnormal cardiac structure were next analyzed. We found that human PKD myocardium was infiltrated by inflammatory macrophage M1 and M2; expression of transforming growth factor (TGF-ß1) and its receptor were upregulated with overexpression of pSmad3 and ß-catenin. Because patients with PKD have an abnormal kidney function that could potentially affect heart structure, we used a heart-specific PKD mouse model to validate that cardiac hypertrophy and fibrosis were independent from polycystic kidney. In summary, our data show that hearts from human PKD were characterized by hypertrophy, interstitial fibrosis, perivascular fibrosis, and conduction system fibrosis with upregulated TGF-ß1 and its receptor. We suggest that such structural abnormalities may predispose to systolic and diastolic cardiac dysfunction in the PKD myocardium.


Subject(s)
Cardiomyopathies , Polycystic Kidney Diseases , Transforming Growth Factor beta1 , Animals , Humans , Mice , Cardiomegaly , Fibrosis , Macrophages/metabolism , Myocardium/metabolism , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/pathology , Transforming Growth Factor beta1/metabolism
6.
Am J Transplant ; 22(2): 565-573, 2022 02.
Article in English | MEDLINE | ID: mdl-34464505

ABSTRACT

Despite the common detection of non-donor specific anti-HLA antibodies (non-DSAs) after lung transplantation, their clinical significance remains unclear. In this retrospective single-center cohort study of 325 lung transplant recipients, we evaluated the association between donor-specific HLA antibodies (DSAs) and non-DSAs with subsequent CLAD development. DSAs were detected in 30% of recipients and were associated with increased CLAD risk, with higher HRs for both de novo and high MFI (>5000) DSAs. Non-DSAs were detected in 56% of recipients, and 85% of DSA positive tests had concurrent non-DSAs. In general, non-DSAs did not increase CLAD risk in multivariable models accounting for DSAs. However, non-DSAs in conjunction with high BAL CXCL9 levels were associated with increased CLAD risk. Multivariable proportional hazards models demonstrate the importance of the HLA antibody-CXCL9 interaction: CLAD risk increases when HLA antibodies (both DSAs and non-DSAs) are detected in conjunction with high CXCL9. Conversely, CLAD risk is not increased when HLA antibodies are detected with low CXCL9. This study supports the potential utility of BAL CXCL9 measurement as a biomarker to risk stratify HLA antibodies for future CLAD. The ability to discriminate between high versus low-risk HLA antibodies may improve management by allowing for guided treatment decisions.


Subject(s)
HLA Antigens , Lung Transplantation , Allografts , Biomarkers , Chemokine CXCL9 , Cohort Studies , Graft Rejection/diagnosis , Graft Rejection/etiology , Graft Survival , Humans , Isoantibodies , Lung Transplantation/adverse effects , Prognosis , Retrospective Studies , Tissue Donors
7.
Am J Transplant ; 22(9): 2169-2179, 2022 09.
Article in English | MEDLINE | ID: mdl-35634722

ABSTRACT

Histopathologic lung allograft injuries are putative harbingers for chronic lung allograft dysfunction (CLAD). However, the mechanisms responsible are not well understood. CXCL9 and CXCL10 are potent chemoattractants of mononuclear cells and potential propagators of allograft injury. We hypothesized that these chemokines would be quantifiable in plasma, and would associate with subsequent CLAD development. In this prospective multicenter study, we evaluated 721 plasma samples for CXCL9/CXCL10 levels from 184 participants at the time of transbronchial biopsies during their first-year post-transplantation. We determined the association between plasma chemokines, histopathologic injury, and CLAD risk using Cox proportional hazards models. We also evaluated CXCL9/CXCL10 levels in bronchoalveolar lavage (BAL) fluid and compared plasma to BAL with respect to CLAD risk. Plasma CXCL9/CXCL10 levels were elevated during the injury patterns associated with CLAD, acute rejection, and acute lung injury, with a dose-response relationship between chemokine levels and CLAD risk. Importantly, there were strong interactions between injury and plasma CXCL9/CXCL10, where histopathologic injury associated with CLAD only in the presence of elevated plasma chemokines. We observed similar associations and interactions with BAL CXCL9/CXCL10 levels. Elevated plasma CXCL9/CXCL10 during allograft injury may contribute to CLAD pathogenesis and has potential as a minimally invasive immune monitoring biomarker.


Subject(s)
Graft vs Host Disease , Lung Transplantation , Allografts , Biomarkers , Chemokine CXCL10 , Chemokine CXCL9 , Graft Rejection/diagnosis , Graft Rejection/etiology , Humans , Lung , Lung Transplantation/adverse effects , Prospective Studies
8.
Catheter Cardiovasc Interv ; 98(1): 139-147, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33825308

ABSTRACT

OBJECTIVES: A large-diameter, intravascular, self-expanding stent system capable of continued expansion during somatic and vascular growth was modeled with finite element analysis (FEA), manufactured and tested in an animal model. BACKGROUND: Children can quickly outgrow intravascular stents. If a stent could expand after implantation in arteries this would be ideal for use in pediatric patients. METHODS: Computer-aided design and FEA were used to design and manufacture large-diameter, self-expanding nitinol stents with both high and low chronic outward force (COF). Four distinct stents with similar designs but with variable lengths and strut thicknesses were manufactured. Fourteen of these stents were implanted in the abdominal aortas or iliac arteries of four juvenile swine. RESULTS: All animals survived without complication to their designated time points of harvest (90 or 180-days), and all stents expanded to greater diameters than the adjacent non-stented artery. Luminal diameter growth was 34-49% and 20-23% for stented and non-stented segments, respectively. Histologic examination revealed variable degrees of the internal elastic lamina and/or medial disruption with a mean injury score ranging from 0.70 ± 0.56 to 1.23 ± 0.21 and low COF stents implanted in smaller arteries having a larger injury score. Inflammatory responses and stenosis formation were minimal and ranged from 0.50 ± 0.71 to 3.00 ± 0.00 and 5.52 ± 1.05% to 14.68 ± 9.12%, respectively. The stent's COF did not correlate with vessel expansion or vascular injury. CONCLUSIONS: Self-expanding stents can mirror and even exceed somatic growth. Although longer-term testing is needed, it may be possible to custom tailor self-expanding stents to expand after arterial implantation in pediatric patients.


Subject(s)
Alloys , Stents , Animals , Biology , Child , Constriction, Pathologic , Humans , Iliac Artery/diagnostic imaging , Iliac Artery/surgery , Swine , Treatment Outcome
9.
Semin Respir Crit Care Med ; 42(3): 392-410, 2021 06.
Article in English | MEDLINE | ID: mdl-34030202

ABSTRACT

The primary factor that limits long-term survival after lung transplantation is chronic lung allograft dysfunction (CLAD). CLAD also impairs quality of life and increases the costs of medical care. Our understanding of CLAD continues to evolve. Consensus definitions of CLAD and the major CLAD phenotypes were recently updated and clarified, but it remains to be seen whether the current definitions will lead to advances in management or impact care. Understanding the potential differences in pathogenesis for each CLAD phenotype may lead to novel therapeutic strategies, including precision medicine. Recognition of CLAD risk factors may lead to earlier interventions to mitigate risk, or to avoid risk factors all together, to prevent the development of CLAD. Unfortunately, currently available therapies for CLAD are usually not effective. However, novel therapeutics aimed at both prevention and treatment are currently under investigation. We provide an overview of the updates to CLAD-related terminology, clinical phenotypes and their diagnosis, natural history, pathogenesis, and potential strategies to treat and prevent CLAD.


Subject(s)
Lung Transplantation , Quality of Life , Allografts , Chronic Disease , Humans , Lung , Lung Transplantation/adverse effects
10.
Am J Transplant ; 20(12): 3308-3318, 2020 12.
Article in English | MEDLINE | ID: mdl-32476272

ABSTRACT

The XVth Banff Conference on Allograft Pathology meeting was held on September 23-27, 2019, in Pittsburgh, Pennsylvania, USA. During this meeting, two main topics in cardiac transplant pathology were addressed: (a) Improvement of endomyocardial biopsy (EMB) accuracy for the diagnosis of rejection and other significant injury patterns, and (b) the orphaned lesion known as Quilty effect or nodular endocardial infiltrates. Molecular technologies have evolved in recent years, deciphering pathophysiology of cardiac rejection. Diagnostically, it is time to integrate the histopathology of EMBs and molecular data. The goal is to incorporate molecular pathology, performed on the same paraffin block as a companion test for histopathology, to yield more accurate and objective EMB interpretation. Application of digital image analysis from hematoxylin and eosin (H&E) stain to multiplex labeling is another means of extracting additional information from EMBs. New concepts have emerged exploring the multifaceted significance of myocardial injury, minimal rejection patterns supported by molecular profiles, and lesions of arteriolitis/vasculitis in the setting of T cell-mediated rejection (TCMR) and antibody-mediated rejection (AMR). The orphaned lesion known as Quilty effect or nodular endocardial infiltrates. A state-of-the-art session with historical aspects and current dilemmas was reviewed, and possible pathogenesis proposed, based on advances in immunology to explain conflicting data. The Quilty effect will be the subject of a multicenter project to explore whether it functions as a tertiary lymphoid organ.


Subject(s)
Graft Rejection , Heart Transplantation , Myocardium , Allografts , Biopsy , Graft Rejection/diagnosis , Graft Rejection/etiology , Heart Transplantation/adverse effects , Humans , Myocardium/pathology , Pennsylvania
11.
Am J Transplant ; 20(10): 2686-2702, 2020 10.
Article in English | MEDLINE | ID: mdl-32320528

ABSTRACT

HLA donor-specific antibodies (DSAs) binding to vascular endothelial cells of the allograft trigger inflammation, vessel injury, and antibody-mediated rejection (AMR). Accumulation of intragraft-recipient macrophages is a histological characteristic of AMR, which portends worse outcome. HLA class I (HLA I) DSAs enhance monocyte recruitment by activating endothelial cells and engaging FcγRs, but the DSA-activated donor endothelial influence on macrophage differentiation is unknown. In this study, we explored the consequence of DSA-activated endothelium on infiltrating monocyte differentiation. Here we show that cardiac allografts from murine recipients treated with MHC I DSA upregulated genes related to monocyte transmigration and Fc receptor stimulation. Human monocytes co-cultured with HLA I IgG-stimulated primary human endothelium promoted monocyte differentiation into CD68+ CD206+ CD163+ macrophages (M(HLA I IgG)), whereas HLA I F(ab')2 stimulated endothelium solely induced higher CD206 (M(HLA I F(ab')2 )). Both macrophage subtypes exhibited significant changes in discrete cytokines/chemokines and unique gene expression profiles. Cross-comparison of gene transcripts between murine DSA-treated cardiac allografts and human co-cultured macrophages identified overlapping genes. These findings uncover the role of HLA I DSA-activated endothelium in monocyte differentiation, and point to a novel, remodeling phenotype of infiltrating macrophages that may contribute to vascular injury.


Subject(s)
Endothelial Cells , Graft Rejection , Allografts , Animals , Graft Rejection/etiology , HLA Antigens , Humans , Inflammation/etiology , Isoantibodies , Macrophages , Mice , Phenotype , Tissue Donors
12.
Am J Transplant ; 20(10): 2768-2780, 2020 10.
Article in English | MEDLINE | ID: mdl-32185871

ABSTRACT

We analyzed humoral immune responses to nonhuman leukocyte antigen (HLA) after cardiac transplantation to identify antibodies associated with allograft rejection. Protein microarray identified 366 non-HLA antibodies (>1.5 fold, P < .5) from a discovery cohort of HLA antibody-negative, endothelial cell crossmatch-positive sera obtained from 12 cardiac allograft recipients at the time of biopsy-proven rejection. From these, 19 plasma membrane proteins and 10 autoantigens identified from gene ontology analysis were combined with 48 proteins identified through literature search to generate a multiplex bead array. Longitudinal sera from a multicenter cohort of adult cardiac allograft recipients (samples: n = 477 no rejection; n = 69 rejection) identified 18 non-HLA antibodies associated with rejection (P < .1) including 4 newly identified non-HLA antigenic targets (DEXI, EMCN, LPHN1, and SSB). CART analysis showed 5/18 non-HLA antibodies distinguished rejection vs nonrejection. Antibodies to 4/18 non-HLA antigens synergize with HLA donor-specific antibodies and significantly increase the odds of rejection (P < .1). The non-HLA panel was validated using an independent adult cardiac transplant cohort (n = 21 no rejection; n = 42 rejection, >1R) with an area under the curve of 0.87 (P < .05) with 92.86% sensitivity and 66.67% specificity. We conclude that multiplex bead array assessment of non-HLA antibodies identifies cardiac transplant recipients at risk of rejection.


Subject(s)
Graft Rejection , Heart Transplantation , Allografts , Antibodies , Graft Rejection/diagnosis , Graft Rejection/etiology , HLA Antigens , Heart Transplantation/adverse effects
13.
J Mol Cell Cardiol ; 129: 154-164, 2019 04.
Article in English | MEDLINE | ID: mdl-30802459

ABSTRACT

OBJECTIVE: To investigate the novel role of Paraoxonase 2 (PON2) in modulating acute myocardial ischemia-reperfusion injury (IRI). APPROACH: IRI was induced both in vivo and ex vivo in male, C57BL6/J (WT) and PON2-deficient (PON-def) mice. In addition, in vitro hypoxia-reoxygenation injury (HRI) was induced in H9c2 cells expressing empty vector (H9c2-EV) or human PON2 (H9c2-hPON2) ±â€¯LY294002 (a potent PI3K inhibitor). Infarct size, PON2 gene expression, mitochondrial calcium retention capacity (CRC), reactive oxygen species (ROS) generation, mitochondrial membrane potential, CHOP and pGSK-3ß protein levels, and cell apoptosis were evaluated. RESULTS: PON2 gene expression is upregulated in WT mice following in vivo IRI. PON2-def mice exhibit a 2-fold larger infarct, increased CHOP levels, and reduced pGSK-3ß levels compared to WT controls. Global cardiac mitochondria isolated from PON2-def mice exhibit reduced CRC and increased ROS production. Cardiomyocytes isolated from PON2-def mice subjected to ex vivo IRI have mitochondria with reduced CRC (also seen under non-IRI conditions), and increased ROS generation and apoptosis compared to WT controls. PON2 knockdown in H9c2 cells subjected to HRI leads to an increase in mitochondrial membrane depolarization. H9c2-hPON2 cells exhibit i) improvement in mitochondrial membrane potential, pGSK-3ß levels and mitochondrial CRC, and ii) decrease in CHOP levels, mitochondrial ROS generation and cell apoptosis, when compared to H9c2-EV controls. Treatment with LY294002 resulted in a decrease of mitochondrial CRC and increase in mitochondrial ROS production and cell apoptosis in the H9c2-hPON2 group versus H9c2-EV controls. CONCLUSION: PON2 protects against acute myocardial IRI by reducing mitochondrial dysfunction and oxidative stress in cardiomyocytes via activation of the PI3K/Akt/GSK-3ß RISK pathway.


Subject(s)
Aryldialkylphosphatase/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Mitochondria, Heart/pathology , Myocardial Reperfusion Injury/prevention & control , Oxidative Stress , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Acute Disease , Animals , Apoptosis , Aryldialkylphosphatase/deficiency , Cardiotonic Agents/metabolism , Cell Line , Humans , Male , Membrane Potential, Mitochondrial , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphorylation , Rats
14.
Curr Cardiol Rep ; 21(8): 81, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31278595

ABSTRACT

PURPOSE OF REVIEW: This discussion is intended to review the anatomy and pathology of the aortic valve and aortic root region, and to provide a basis for the understanding of and treatment of the important life-threatening diseases that affect the aortic valve. RECENT FINDINGS: The most exciting recent finding is that less invasive methods are being developed to treat diseases of the aortic valve. There are no medical cures for aortic valve diseases. Until recently, open-heart surgery was the only effective method of treatment. Now percutaneous approaches to implant bioprosthetic valves into failed native or previously implanted bioprosthetic valves are being developed and utilized. A genetic basis for many of the diseases that affect the aortic valve is being discovered that also should lead to innovative approaches to perhaps prevent these disease. Sequencing of ribosomal RNA is assisting in identifying organisms causing endocarditis, leading to more effective antimicrobial therapy. There is exciting, expanding, therapeutic innovation in the treatment of aortic valve disease.


Subject(s)
Aortic Valve Insufficiency/pathology , Aortic Valve Stenosis/pathology , Aortic Valve/pathology , Heart Valve Prosthesis , Humans , Transcatheter Aortic Valve Replacement , Treatment Outcome
15.
Curr Cardiol Rep ; 21(7): 54, 2019 05 18.
Article in English | MEDLINE | ID: mdl-31104143

ABSTRACT

PURPOSE OF REVIEW: This review describes the normal structure and pathologic changes that affect the right-sided cardiac valves and chambers. RECENT FINDINGS: The anatomy and pathology described have been known for many years. Knowledge of these findings has gained relevance. The pattern of endocarditis is changing. New diagnostic techniques have allowed better characterization of lesions responsible for cardiac dysfunction. Novel, less invasive interventions have made recognition of abnormalities more clinically relevant. There are many different pathologic entities that can affect the right-sided cardiac valves. These are discussed in this review.


Subject(s)
Pulmonary Valve , Tricuspid Valve Insufficiency , Humans
16.
Curr Cardiol Rep ; 21(7): 61, 2019 05 23.
Article in English | MEDLINE | ID: mdl-31123911

ABSTRACT

PURPOSE OF REVIEW: This review describes numerous pathologic entities that cause structural abnormalities of the mitral valve. Different pathologic entities involve different components of the so-called mitral apparatus: atrial wall, annulus, leaflets, chordae, papillary muscles, and/or left ventricular free wall. These abnormalities can cause valvular stenosis, regurgitation, or both. RECENT FINDINGS: Currently, in addition to open-chest surgery to replace or repair the damaged mitral valve, there are less invasive percutaneous approaches to address mitral valve dysfunction. These include narrowing the orifice, clipping the leaflets, and inserting bioprostheses percutaneously. Understanding the structural abnormalities discussed in this review is essential for choosing the optimal therapeutic intervention for mitral valve disease.


Subject(s)
Bioprosthesis , Heart Valve Diseases/pathology , Mitral Valve Insufficiency/pathology , Mitral Valve/abnormalities , Heart Atria , Heart Valve Diseases/therapy , Humans , Mitral Valve/pathology , Mitral Valve Insufficiency/therapy
17.
Curr Cardiol Rep ; 21(7): 63, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31147795

ABSTRACT

PURPOSE OF REVIEW: This review discusses the normal anatomy and pathology of the tricuspid valve (TV) and right side of the heart. Emphasis is on those anatomic and pathologic features relevant to interventions intended to restore normal function to the TV in disease states. RECENT FINDINGS: TV pathology is less common than aortic and mitral valve pathology, and treatment and outcomes for interventions face considerable hurdles. New innovations and early data showing safety and efficacy in transcatheter interventions have transformed TV interventions into the next frontier in cardiac valve disease treatment. Certain features of the TV and right heart have presented themselves as potential targets, as well as impediments, for TV intervention. The causes of TV pathology and the anatomy of the TV and right heart bring unique challenges to intervention. Approaches to intervention will continue to progress and change the way we view and treat TV pathology.


Subject(s)
Heart Valve Diseases/pathology , Tricuspid Valve Insufficiency/pathology , Tricuspid Valve/anatomy & histology , Tricuspid Valve/pathology , Heart Valve Prosthesis Implantation , Humans , Treatment Outcome , Tricuspid Valve/surgery , Tricuspid Valve Insufficiency/surgery
18.
BMC Med Genet ; 19(1): 1, 2018 01 03.
Article in English | MEDLINE | ID: mdl-29298659

ABSTRACT

BACKGROUND: The Protein Kinase AMP-Activated Non-Catalytic Subunit Gamma 2 (PRKAG2) cardiac syndrome is characterized by glycogen accumulation in the cardiac tissue. The disease presents clinically with hypertrophic cardiomyopathy (HCM), and it is often associated with conduction abnormalities. CASE PRESENTATION: A 23 year-old female with history of Wolff-Parkinson-White (WPW) and HCM presented for evaluation after an episode of Non-ST Elevation Myocardial Infarction (NSTEMI). The patient was found to have severe coronary bridging on angiography and underwent an unroofing of the left anterior descending artery (LAD). Due to the constellation of symptoms, the patient underwent genetic testing and a cardiac muscle biopsy. Genetic testing was significant for an Arg302Gln mutation in the PRKAG2 gene. Cardiac tissue biopsy revealed significant myocyte hypertrophy and large vacuoles with glycogen stores. CONCLUSION: The pathologic and genetics findings of our patient are consistent with PRKAG2 syndrome. Patients presenting with conduction abnormalities and suspected HCM should be considered for genetic testing to identify possible underlying genetic etiologies.


Subject(s)
AMP-Activated Protein Kinases/genetics , Cardiomyopathy, Hypertrophic/genetics , Non-ST Elevated Myocardial Infarction/genetics , Wolff-Parkinson-White Syndrome/genetics , Angiography , Biopsy , Cardiomyopathy, Hypertrophic/diagnosis , Chromosomes, Human, Pair 7/genetics , Female , Genetic Testing , Heart/physiopathology , Humans , Mutation , Myocardium/pathology , Non-ST Elevated Myocardial Infarction/diagnosis , Wolff-Parkinson-White Syndrome/diagnosis , Young Adult
19.
Catheter Cardiovasc Interv ; 91(2): 277-284, 2018 02 01.
Article in English | MEDLINE | ID: mdl-28895275

ABSTRACT

OBJECTIVES: To provide a comparison of the outcome of infective endocarditis (IE) in patients undergoing transcatheter pulmonary valve replacement (TPVR) versus surgical pulmonary valve replacement (SPVR). BACKGROUND: Although TPVR is thought to be associated with a higher risk of IE than SPVR, there is paucity of data to support this. METHODS: Patients who underwent TPVR or SPVR at UCLA between October 2010 and September 2016 were included and retrospectively analyzed. RESULTS: Three hundred forty-two patients underwent PVR at UCLA including 134 SPVR and 208 TPVR. Patients undergoing TPVR were more likely to have had a history of endocarditis than those undergoing SPVR (5.3% vs. 0.7%, P = 0.03) and a right ventricle to pulmonary artery (RV to PA) conduit (37% vs. 17%, P = 0.0001). Two SPVR and seven TPVR patients developed IE with a 4-year freedom from endocarditis of 94.0% in the SPVR versus 84% in the TPVR group (P = 0.13). In patients who underwent TPVR and developed endocarditis, the mean gradient across the RVOT prior to intervention was higher (28.1 ± 4.5 vs. 17.4 ± 0.6 mmHg, P = 0.02) and were more likely to have a conduit (71% vs. 36%, P = 0.049). CONCLUSIONS: In this study, patients undergoing TPVR were not at a higher risk of IE than patients undergoing SPVR. TPVR patients were more likely to have had a prior history of IE and RV-PA conduit. The patients at highest risk were those with stenotic RV to PA conduits who were treated with TPVR.


Subject(s)
Cardiac Catheterization/adverse effects , Endocarditis/epidemiology , Heart Valve Prosthesis Implantation/adverse effects , Pulmonary Valve/surgery , Adolescent , Adult , Cardiac Catheterization/instrumentation , Cardiac Catheterization/methods , Child , Endocarditis/diagnosis , Endocarditis/therapy , Female , Heart Valve Prosthesis Implantation/instrumentation , Heart Valve Prosthesis Implantation/methods , Humans , Incidence , Los Angeles/epidemiology , Male , Pulmonary Valve/diagnostic imaging , Pulmonary Valve/physiopathology , Retrospective Studies , Risk Factors , Time Factors , Treatment Outcome , Young Adult
20.
Semin Respir Crit Care Med ; 39(4): 425-433, 2018 08.
Article in English | MEDLINE | ID: mdl-30404110

ABSTRACT

Vasculitides are a heterogeneous group of disorders in which inflammation of blood vessel walls is present at least some time during the course of the disease. Vasculitides can affect any caliber or type of vessel in many anatomic sites; however, the disease can alter more than just vasculature. Given the diversity of vasculitides, in 2012, a revised classification system was proposed to categorize vasculitides by the type of vessel involved including size, function, and structural attributes. In the lung, vasculitis impacts both the pulmonary vessels and parenchyma. Extrapulmonary involvement, particularly with concomitant kidney involvement, is a frequent occurrence. Pulmonary vasculitides often present with hemoptysis, pathologically manifested as diffuse alveolar hemorrhage (DAH) with or without capillaritis and can be life threatening when diffuse throughout the lungs. Etiologies for DAH include both primary and secondary vasculitides, along with collagen-vascular diseases, infection, and drug toxicity. Therefore, diagnosing the specific vasculitic etiology often relies on comprehensive assessment of all clinical, laboratory/serological, imaging, and histopathologic features that may be present. The most common primary pulmonary vasculitides often affect small vessels and are associated with circulating antineutrophilic cytoplasmic antibodies (ANCAs). In the 2012 classification, these include granulomatosis with polyangiitis (formerly Wegener granulomatosis), eosinophilic granulomatosis with polyangiitis (formerly Churg-Strauss' syndrome), and microscopic polyangiitis. Other less frequent vasculitides that are non-ANCA associated or affect medium- to large-sized vessels can have pulmonary involvement. These entities are usually associated with extrapulmonary disease and include polyarteritis nodosa, Takayasu's arteritis, Behçet's disease, and antibasement membrane antibody disease (formerly Goodpasture's syndrome). Although all vasculitides have vessel wall inflammation at some phase in the disease process, their histopathologic findings are as diverse as the group of diseases themselves. The characteristic histologic findings of the pulmonary vasculitides will be reviewed here.


Subject(s)
Hemorrhage/etiology , Pulmonary Alveoli/pathology , Vasculitis/pathology , Antibodies, Antineutrophil Cytoplasmic/blood , Churg-Strauss Syndrome/complications , Churg-Strauss Syndrome/pathology , Granulomatosis with Polyangiitis/complications , Granulomatosis with Polyangiitis/pathology , Humans , Microscopic Polyangiitis/complications , Microscopic Polyangiitis/pathology , Vasculitis/complications
SELECTION OF CITATIONS
SEARCH DETAIL