Your browser doesn't support javascript.
loading
Early Injury Landscape in Vein Harvest by Single-Cell and Spatial Transcriptomics.
Michaud, Marina E; Mota, Lucas; Bakhtiari, Mojtaba; Thomas, Beena E; Tomeo, John; Pilcher, William; Contreras, Mauricio; Ferran, Christiane; Bhasin, Swati S; Pradhan-Nabzdyk, Leena; LoGerfo, Frank W; Liang, Patric; Bhasin, Manoj K.
  • Michaud ME; Department of Pediatrics, Emory School of Medicine, Atlanta, GA (M.E.M., M.B., B.E.T., S.S.B., M.K.B.).
  • Mota L; Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center (L.M., J.T., M.C., C.F., L.P.-N., F.W.L., P.L.), Harvard Medical School, Boston, MA.
  • Bakhtiari M; Department of Pediatrics, Emory School of Medicine, Atlanta, GA (M.E.M., M.B., B.E.T., S.S.B., M.K.B.).
  • Thomas BE; Department of Pediatrics, Emory School of Medicine, Atlanta, GA (M.E.M., M.B., B.E.T., S.S.B., M.K.B.).
  • Tomeo J; Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center (L.M., J.T., M.C., C.F., L.P.-N., F.W.L., P.L.), Harvard Medical School, Boston, MA.
  • Pilcher W; Department of Biomedical Engineering, Emory University, Atlanta, GA (W.P., M.K.B.).
  • Contreras M; Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center (L.M., J.T., M.C., C.F., L.P.-N., F.W.L., P.L.), Harvard Medical School, Boston, MA.
  • Ferran C; Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center (L.M., J.T., M.C., C.F., L.P.-N., F.W.L., P.L.), Harvard Medical School, Boston, MA.
  • Bhasin SS; Department of Medicine, Beth Israel Deaconess Medical Center, Center for Vascular Biology Research and the Division of Nephrology (C.F.), Harvard Medical School, Boston, MA.
  • Pradhan-Nabzdyk L; Department of Pediatrics, Emory School of Medicine, Atlanta, GA (M.E.M., M.B., B.E.T., S.S.B., M.K.B.).
  • LoGerfo FW; Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, GA (S.S.B., M.K.B.).
  • Liang P; Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center (L.M., J.T., M.C., C.F., L.P.-N., F.W.L., P.L.), Harvard Medical School, Boston, MA.
  • Bhasin MK; Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center (L.M., J.T., M.C., C.F., L.P.-N., F.W.L., P.L.), Harvard Medical School, Boston, MA.
Circ Res ; 135(1): 110-134, 2024 Jun 21.
Article en En | MEDLINE | ID: mdl-38808504
ABSTRACT

BACKGROUND:

Vein graft failure following cardiovascular bypass surgery results in significant patient morbidity and cost to the healthcare system. Vein graft injury can occur during autogenous vein harvest and preparation, as well as after implantation into the arterial system, leading to the development of intimal hyperplasia, vein graft stenosis, and, ultimately, bypass graft failure. Although previous studies have identified maladaptive pathways that occur shortly after implantation, the specific signaling pathways that occur during vein graft preparation are not well defined and may result in a cumulative impact on vein graft failure. We, therefore, aimed to elucidate the response of the vein conduit wall during harvest and following implantation, probing the key maladaptive pathways driving graft failure with the overarching goal of identifying therapeutic targets for biologic intervention to minimize these natural responses to surgical vein graft injury.

METHODS:

Employing a novel approach to investigating vascular pathologies, we harnessed both single-nuclei RNA-sequencing and spatial transcriptomics analyses to profile the genomic effects of vein grafts after harvest and distension, then compared these findings to vein grafts obtained 24 hours after carotid-carotid vein bypass implantation in a canine model (n=4).

RESULTS:

Spatial transcriptomic analysis of canine cephalic vein after initial conduit harvest and distention revealed significant enrichment of pathways (P<0.05) involved in the activation of endothelial cells (ECs), fibroblasts, and vascular smooth muscle cells, namely pathways responsible for cellular proliferation and migration and platelet activation across the intimal and medial layers, cytokine signaling within the adventitial layer, and ECM (extracellular matrix) remodeling throughout the vein wall. Subsequent single-nuclei RNA-sequencing analysis supported these findings and further unveiled distinct EC and fibroblast subpopulations with significant upregulation (P<0.05) of markers related to endothelial injury response and cellular activation of ECs, fibroblasts, and vascular smooth muscle cells. Similarly, in vein grafts obtained 24 hours after arterial bypass, there was an increase in myeloid cell, protomyofibroblast, injury response EC, and mesenchymal-transitioning EC subpopulations with a concomitant decrease in homeostatic ECs and fibroblasts. Among these markers were genes previously implicated in vein graft injury, including VCAN, FBN1, and VEGFC, in addition to novel genes of interest, such as GLIS3 and EPHA3. These genes were further noted to be driving the expression of genes implicated in vascular remodeling and graft failure, such as IL-6, TGFBR1, SMAD4, and ADAMTS9. By integrating the spatial transcriptomics and single-nuclei RNA-sequencing data sets, we highlighted the spatial architecture of the vein graft following distension, wherein activated and mesenchymal-transitioning ECs, myeloid cells, and fibroblasts were notably enriched in the intima and media of distended veins. Finally, intercellular communication network analysis unveiled the critical roles of activated ECs, mesenchymal-transitioning ECs, protomyofibroblasts, and vascular smooth muscle cells in upregulating signaling pathways associated with cellular proliferation (MDK [midkine], PDGF [platelet-derived growth factor], VEGF [vascular endothelial growth factor]), transdifferentiation (Notch), migration (ephrin, semaphorin), ECM remodeling (collagen, laminin, fibronectin), and inflammation (thrombospondin), following distension.

CONCLUSIONS:

Vein conduit harvest and distension elicit a prompt genomic response facilitated by distinct cellular subpopulations heterogeneously distributed throughout the vein wall. This response was found to be further exacerbated following vein graft implantation, resulting in a cascade of maladaptive gene regulatory networks. Together, these results suggest that distension initiates the upregulation of pathological pathways that may ultimately contribute to bypass graft failure and presents potential early targets warranting investigation for targeted therapies. This work highlights the first applications of single-nuclei and spatial transcriptomic analyses to investigate venous pathologies, underscoring the utility of these methodologies and providing a foundation for future investigations.
Asunto(s)
Palabras clave

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Análisis de la Célula Individual / Transcriptoma Límite: Animals Idioma: En Año: 2024 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Análisis de la Célula Individual / Transcriptoma Límite: Animals Idioma: En Año: 2024 Tipo del documento: Article