Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jeans M. Santana is active.

Publication


Featured researches published by Jeans M. Santana.


Physiological Reports | 2016

Pericardial patch venoplasty heals via attraction of venous progenitor cells

Hualong Bai; Mo Wang; Trenton R. Foster; Haidi Hu; Hao He; Takuya Hashimoto; Jesse J. Hanisch; Jeans M. Santana; Ying Xing; Alan Dardik

Pericardial patches are commonly used during cardiovascular surgery to close blood vessels. In arteries, patches accumulate arterial progenitor cells; we hypothesized that venous patches would accumulate venous progenitor cells, in the absence of arterial pressure. We developed a novel rat inferior vena cava (IVC) venotomy model and repaired it with a pericardial patch. Cells infiltrated the patch to form a thick neointima by day 7; some cells were CD34+/VEGFR2+ and CD31+/Eph‐B4+ consistent with development of venous identity in the healing patch. Compared to arterial patches, the venous patches had increased neointimal thickness at day 7 without any pseudoaneurysms. Addition of an arteriovenous fistula (AVF) to increase blood flow on the patch resulted in reduced patch neointimal thickness and proliferation, but neointimal thickness was not reversible with AVF ligation. These results show that rat patch venoplasty is a novel model of aggressive venous neointimal hyperplasia.


Journal of Biological Chemistry | 2018

CLOCK phosphorylation by AKT regulates its nuclear accumulation and circadian gene expression in peripheral tissues

Amelia K. Luciano; Wenping Zhou; Jeans M. Santana; Cleo Kyriakides; Heino Velazquez; William C. Sessa

Circadian locomotor output cycles kaput (CLOCK) is a transcription factor that activates transcription of clock-controlled genes by heterodimerizing with BMAL1 and binding to E-box elements on DNA. Although several phosphorylation sites on CLOCK have already been identified, this study characterizes a novel phosphorylation site at serine 845 (Ser-836 in humans). Here, we show that CLOCK is a novel AKT substrate in vitro and in cells, and this phosphorylation site is a negative regulator of CLOCK nuclear localization by acting as a binding site for 14-3-3 proteins. To examine the role of CLOCK phosphorylation in vivo, ClockS845A knockin mice were generated using CRISPR/Cas9 technology. ClockS845A mice are essentially normal with normal central circadian rhythms and hemodynamics. However, examination of core circadian gene expression from peripheral tissues demonstrated that ClockS845A mice have diminished expression of Per2, Reverba, Dbp, and Npas2 in skeletal muscle and Per2, Reverba, Dbp, Per1, Rora, and Npas2 in the liver during the circadian cycle. The reduction in Dbp levels is associated with reduced H3K9ac at E-boxes where CLOCK binds despite no change in total CLOCK levels. Thus, CLOCK phosphorylation by AKT on Ser-845 regulates its nuclear translocation and the expression levels of certain core circadian genes in insulin-sensitive tissues.


Seminars in Vascular Surgery | 2016

Future research directions to improve fistula maturation and reduce access failure

Haidi Hu; Sandeep Patel; Jesse J. Hanisch; Jeans M. Santana; Takuya Hashimoto; Hualong Bai; Tambudzai Kudze; Trenton R. Foster; Jianming Guo; Bogdan Yatsula; Janice Tsui; Alan Dardik

With the increasing prevalence of end-stage renal disease, there is a growing need for hemodialysis. Arteriovenous fistulae (AVF) are the preferred type of vascular access for hemodialysis, but maturation and failure continue to present significant barriers to successful fistula use. AVF maturation integrates outward remodeling with vessel wall thickening in response to drastic hemodynamic changes in the setting of uremia, systemic inflammation, oxidative stress, and pre-existent vascular pathology. AVF can fail due to both failure to mature adequately to support hemodialysis and development of neointimal hyperplasia that narrows the AVF lumen, typically near the fistula anastomosis. Failure due to neointimal hyperplasia involves vascular cell activation and migration and extracellular matrix remodeling with complex interactions of growth factors, adhesion molecules, inflammatory mediators, and chemokines, all of which result in maladaptive remodeling. Different strategies have been proposed to prevent and treat AVF failure based on current understanding of the modes and pathology of access failure; these approaches range from appropriate patient selection and use of alternative surgical strategies for fistula creation, to the use of novel interventional techniques or drugs to treat failing fistulae. Effective treatments to prevent or treat AVF failure require a multidisciplinary approach involving nephrologists, vascular surgeons, and interventional radiologists, careful patient selection, and the use of tailored systemic or localized interventions to improve patient-specific outcomes. This review provides contemporary information on the underlying mechanisms of AVF maturation and failure and discusses the broad spectrum of options that can be tailored for specific therapy.


Birth Defects Research Part C-embryo Today-reviews | 2016

Membrane-mediated regulation of vascular identity.

Takuya Hashimoto; Masayuki Tsuneki; Trenton R. Foster; Jeans M. Santana; Hualong Bai; Mo Wang; Haidi Hu; Jesse J. Hanisch; Alan Dardik

Vascular diseases span diverse pathology, but frequently arise from aberrant signaling attributed to specific membrane-associated molecules, particularly the Eph-ephrin family. Originally recognized as markers of embryonic vessel identity, Eph receptors and their membrane-associated ligands, ephrins, are now known to have a range of vital functions in vascular physiology. Interactions of Ephs with ephrins at cell-to-cell interfaces promote a variety of cellular responses such as repulsion, adhesion, attraction, and migration, and frequently occur during organ development, including vessel formation. Elaborate coordination of Eph- and ephrin-related signaling among different cell populations is required for proper formation of the embryonic vessel network. There is growing evidence supporting the idea that Eph and ephrin proteins also have postnatal interactions with a number of other membrane-associated signal transduction pathways, coordinating translation of environmental signals into cells. This article provides an overview of membrane-bound signaling mechanisms that define vascular identity in both the embryo and the adult, focusing on Eph- and ephrin-related signaling. We also discuss the role and clinical significance of this signaling system in normal organ development, neoplasms, and vascular pathologies.


Journal of Vascular Surgery | 2017

Eph-B4 mediates vein graft adaptation by regulation of endothelial nitric oxide synthase

Mo Wang; Michael J. Collins; Trenton R. Foster; Hualong Bai; Takuya Hashimoto; Jeans M. Santana; Chang Shu; Alan Dardik

Objective: Vein graft adaptation is characterized by loss of expression of the tyrosine kinase receptor Eph‐B4, the embryonic determinant of venous identity, without increased expression of its ligand ephrin‐B2, the embryonic determinant of arterial identity. Endothelial nitric oxide synthase (eNOS) is an important mediator of vessel remodeling. We hypothesized that the mechanism of action of Eph‐B4 during vein graft adaptation might be through regulation of downstream eNOS activity. Methods: Mouse lung endothelial cells were stimulated with ephrin‐B2/Fc, without and with preclustering, without and with the eNOS inhibitor N&ohgr;‐nitro‐L‐arginine methyl ester hydrochloride or the Eph‐B4 inhibitor NVP‐BHG712, and assessed by Western blot and immunofluorescence for eNOS and Eph‐B4 phosphorylation. Nitric oxide (NO) production was assessed using an NO‐specific chemiluminescence analyzer. Cell migration was assessed using a Transwell assay. Human and mouse vein graft specimens were examined for eNOS activity by Western blot, and vessel remodeling was assessed in vein grafts in wild‐type or eNOS knockout mice. Results: Ephrin‐B2/Fc stimulated both Eph‐B4 and eNOS phosphorylation in a bimodal temporal distribution (n = 4; P < .05), with preclustered ephrin‐B2/Fc causing prolonged peak Eph‐B4 and eNOS phosphorylation as well as altered subcellular localization (n = 4; P < .05). Ephrin‐B2/Fc increased NO release (n = 3; P < .01) as well as increased endothelial cell migration (n = 6; P < .05) in an eNOS‐dependent fashion. Both human and mouse vein grafts showed increased eNOS phosphorylation compared with normal veins (n = 3; P < .05). Vein grafts from eNOS knockout mice showed less dilation and less wall thickening compared with wild‐type vein grafts (n = 7; P < .05). Conclusions: eNOS is a mediator of vein graft adaptation to the arterial environment. Eph‐B4 stimulates eNOS phosphorylation in vitro and may mediate vein graft adaptation by regulation of eNOS activity in vivo. Clinical Relevance: To date, despite a large number of clinical trials, no strategy appears effective in improving long‐term vein graft patency. Eph‐B4 activity regulates vein graft adaptation, and this study shows that Eph‐B4 stimulates endothelial nitric oxide synthase (eNOS) phosphorylation, suggesting that Eph‐B4 activity during vein graft adaptation is mediated by eNOS. Modulation of eNOS activity may provide a potential new therapeutic target to improve vein graft patency.


Birth Defects Research Part A-clinical and Molecular Teratology | 2016

Membrane‐mediated regulation of vascular identity

Takuya Hashimoto; Masayuki Tsuneki; Trenton R. Foster; Jeans M. Santana; Hualong Bai; Mo Wang; Haidi Hu; Jesse J. Hanisch; Alan Dardik

Vascular diseases span diverse pathology, but frequently arise from aberrant signaling attributed to specific membrane-associated molecules, particularly the Eph-ephrin family. Originally recognized as markers of embryonic vessel identity, Eph receptors and their membrane-associated ligands, ephrins, are now known to have a range of vital functions in vascular physiology. Interactions of Ephs with ephrins at cell-to-cell interfaces promote a variety of cellular responses such as repulsion, adhesion, attraction, and migration, and frequently occur during organ development, including vessel formation. Elaborate coordination of Eph- and ephrin-related signaling among different cell populations is required for proper formation of the embryonic vessel network. There is growing evidence supporting the idea that Eph and ephrin proteins also have postnatal interactions with a number of other membrane-associated signal transduction pathways, coordinating translation of environmental signals into cells. This article provides an overview of membrane-bound signaling mechanisms that define vascular identity in both the embryo and the adult, focusing on Eph- and ephrin-related signaling. We also discuss the role and clinical significance of this signaling system in normal organ development, neoplasms, and vascular pathologies.


Scientific Reports | 2017

Eph-B4 regulates adaptive venous remodeling to improve arteriovenous fistula patency

Clinton D. Protack; Trenton R. Foster; Takuya Hashimoto; Kota Yamamoto; Monica Y. Lee; Jan R. Kraehling; Hualong Bai; Haidi Hu; Toshihiko Isaji; Jeans M. Santana; Mo Wang; William C. Sessa; Alan Dardik

Low rates of arteriovenous fistula (AVF) maturation prevent optimal fistula use for hemodialysis; however, the mechanism of venous remodeling in the fistula environment is not well understood. We hypothesized that the embryonic venous determinant Eph-B4 mediates AVF maturation. In human AVF and a mouse aortocaval fistula model, Eph-B4 protein expression increased in the fistula vein; expression of the arterial determinant Ephrin-B2 also increased. Stimulation of Eph-B-mediated signaling with Ephrin-B2/Fc showed improved fistula patency with less wall thickness. Mutagenesis studies showed that tyrosine-774 is critical for Eph-B4 signaling and administration of inactive Eph-B4-Y774F increased fistula wall thickness. Akt1 expression also increased in AVF; Akt1 knockout mice showed reduced fistula diameter and wall thickness. In Akt1 knockout mice, stimulation of Eph-B signaling with Ephrin-B2/Fc showed no effect on remodeling. These results show that AVF maturation is associated with acquisition of dual arteriovenous identity; increased Eph-B activity improves AVF patency. Inhibition of Akt1 function abolishes Eph-B-mediated venous remodeling suggesting that Eph-B4 regulates AVF venous adaptation through an Akt1-mediated mechanism.


Journal of Visualized Experiments | 2017

Patch Angioplasty in the Rat Aorta or Inferior Vena Cava

Hualong Bai; Xin Li; Takuya Hashimoto; Haidi Hu; Trenton R. Foster; Jesse J. Hanisch; Jeans M. Santana; Alan Dardik

Pericardial patches are commonly used in vascular surgery to close vessels. To facilitate studies of the neointimal hyperplasia that forms on the patch, we developed a rat model of patch angioplasty that can be used in either a vein or an artery, creating a patch venoplasty or a patch arterioplasty, respectively. Technical aspects of this model are discussed. The infra-renal IVC or aorta are dissected and then clamped proximally and distally. A 3 mm venotomy or arteriotomy is performed in the infrarenal inferior vena cava or aorta of 6 to 8 week-old Wistar rats. A bovine pericardial patch (3 mm x 1.5 mm x 0.6 mm) is then used to close the site using a 10-0 nylon suture. Compared to arterial patches, venous patches show increased neointimal thickness on postoperative day 7. This novel model of pericardial patch angioplasty can be used to examine neointimal hyperplasia on vascular biomaterials, as well as to compare the differences between the arterial and venous environments.


Annals of Vascular Diseases | 2017

Improving the Outcome of Vein Grafts: Should Vascular Surgeons Turn Veins into Arteries?

Toshihiko Isaji; Takuya Hashimoto; Kota Yamamoto; Jeans M. Santana; Bogdan Yatsula; Haidi Hu; Hualong Bai; Guo Jianming; Tambudzai Kudze; Toshiya Nishibe; Alan Dardik

Autogenous vein grafts remain the gold standard conduit for arterial bypass, particularly for the treatment of critical limb ischemia. Vein graft adaptation to the arterial environment, i.e., adequate dilation and wall thickening, contributes to the superior performance of vein grafts. However, abnormal venous wall remodeling with excessive neointimal hyperplasia commonly causes vein graft failure. Since the PREVENT trials failed to improve vein graft outcomes, new strategies focus on the adaptive response of the venous endothelial cells to the post-surgical arterial environment. Eph-B4, the determinant of venous endothelium during embryonic development, remains expressed and functional in adult venous tissue. After surgery, vein grafts lose their venous identity, with loss of Eph-B4 expression; however, arterial identity is not gained, consistent with loss of all vessel identity. In mouse vein grafts, stimulation of venous Eph-B4 signaling promotes retention of venous identity in endothelial cells and is associated with vein graft walls that are not thickened. Eph-B4 regulates downstream signaling pathways of relevance to vascular biology, including caveolin-1, Akt, and endothelial nitric oxide synthase (eNOS). Regulation of the Eph-B4 signaling pathway may be a novel therapeutic target to prevent vein graft failure.


Journal of Biological Rhythms | 2017

Akt1 Controls the Timing and Amplitude of Vascular Circadian Gene Expression

Amelia K. Luciano; Jeans M. Santana; Heino Velazquez; William C. Sessa

The AKT signaling pathway is important for circadian rhythms in mammals and flies (Drosophila). However, AKT signaling in mammals is more complicated since there are 3 isoforms of AKT, each performing slightly different functions. Here we study the most ubiquitous AKT isoform, Akt1, and its role at the organismal level in the central and vascular peripheral clocks. Akt1–/– mice exhibit relatively normal behavioral rhythms with only minor differences in circadian gene expression in the liver and heart. However, circadian gene expression in the Akt1–/– aorta, compared with control aorta, follows a distinct pattern. In the Akt1–/– aorta, positive regulators of circadian transcription have lower amplitude rhythms and peak earlier in the day, and negative circadian regulators are expressed at higher amplitudes and peak later in the day. In endothelial cells, negative circadian regulators exhibit an increased amplitude of expression, while the positive circadian regulators are arrhythmic with a decreased amplitude of expression. This indicates that Akt1 conditions the normal circadian rhythm in the vasculature more so than in other peripheral tissues where other AKT isoforms or kinases might be important for daily rhythms.

Collaboration


Dive into the Jeans M. Santana's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge