Abstract 19641: Novel Microprinted Elisa Platform for High Throughput Screening of Protein Secretion Reveals a Comprehensive Strategy to Prevent Ischemia Reperfusion Induced Apoptosis

Circulation ◽  
2014 ◽  
Vol 130 (suppl_2) ◽  
Author(s):  
Kshitiz Kz ◽  
David E Ellison ◽  
Junaid Afzal ◽  
Maimon E Hubbi ◽  
Segun Bernard ◽  
...  

Introduction: A major challenge in using stem cells to treat myocardial infarction (MI) is the massive cell death post transplant. Many progenitor cell types limit disrepair post MI without significant cardiac differentiation, possibly by anti-apoptotic signals in their secretions. The signature of these secretions is however not well known. Methods: We created a novel high throughput microELISA platform by combining microprinting and microfluidics to precisely estimate secretions of a small population of cells over time. Screening the secretions in many stem cell types used previously to treat MI, i.e. BMSCs (Bone marrow derived stem cells), CDCs (Cardiosphere derived cells), and iPSC-CMs (induced pluripotent stem cell derived cardiomyocytes), we found a common preserved secretory signature of growth factors. Results: Using a high throughput screen of pro-apoptotic factors that prevent CDCs from peroxide induced cell death, we surprisingly found that these factors were identical to the preserved secretory signature. Using the constituents of the anti-apoptotic secretory signature in combination with ischemic and mechanical preconditioning in myocardium mimicking rigidity, we created a comprehensive cytoprotective cocktail to prevent CDCs from ischemia induced cell death. We tested the cocktail in a rat model of ischemia reperfusion, and found a stark reduction in CDC retention post injection. Figures: (A) MicroELISA schematic. Secretions from cells in left are detected by rows of microprinted capture antibodies; photograph in (C-D). Bioluminescence imaging of CDCs treated with comprehensive cocktail show significantly high cell retention vs untreated CDCs.

2021 ◽  
Vol 22 (15) ◽  
pp. 7946
Author(s):  
Chang Youn Lee ◽  
Seahyoung Lee ◽  
Seongtae Jeong ◽  
Jiyun Lee ◽  
Hyang-Hee Seo ◽  
...  

The acute demise of stem cells following transplantation significantly compromises the efficacy of stem cell-based cell therapeutics for infarcted hearts. As the stem cells transplanted into the damaged heart are readily exposed to the hostile environment, it can be assumed that the acute death of the transplanted stem cells is also inflicted by the same environmental cues that caused massive death of the host cardiac cells. Pyroptosis, a highly inflammatory form of programmed cell death, has been added to the list of important cell death mechanisms in the damaged heart. However, unlike the well-established cell death mechanisms such as necrosis or apoptosis, the exact role and significance of pyroptosis in the acute death of transplanted stem cells have not been explored in depth. In the present study, we found that M1 macrophages mediate the pyroptosis in the ischemia/reperfusion (I/R) injured hearts and identified miRNA-762 as an important regulator of interleukin 1b production and subsequent pyroptosis. Delivery of exogenous miRNA-762 prior to transplantation significantly increased the post-transplant survival of stem cells and also significantly ameliorated cardiac fibrosis and heart functions following I/R injury. Our data strongly suggest that suppressing pyroptosis can be an effective adjuvant strategy to enhance the efficacy of stem cell-based therapeutics for diseased hearts.


2020 ◽  
Vol 335 ◽  
pp. 108627 ◽  
Author(s):  
Madel Durens ◽  
Jonathan Nestor ◽  
Madeline Williams ◽  
Kevin Herold ◽  
Robert F. Niescier ◽  
...  

2020 ◽  
Author(s):  
Valentin Parat ◽  
Brigitte Onteniente ◽  
Julien Maruotti

AbstractIn this study, we describe a simple and straight-forward assay using induced pluripotent stem cell derived melanocytes and high-throughput flow cytometry, to screen and identify pigment regulating agents. The assays is based on the correlation between forward light-scatter characteristics and melanin content, with pigmented cells displaying high light absorption/low forward light-scatter, while the opposite is true for lowly pigmented melanocytes, as a result of genetic background or chemical treatments. Orthogonal validation is then performed by regular melanin quantification. Such approach was validated using a set of 80 small molecules, and yielded a confirmed hit. The assay described in this study may prove a useful tool to identify modulators of melanogenesis in human melanocytes.


2020 ◽  
Author(s):  
Jeremy W. Linsley ◽  
Drew A. Linsley ◽  
Josh Lamstein ◽  
Gennadi Ryan ◽  
Kevan Shah ◽  
...  

AbstractCell death is an essential process in biology that must be accounted for in live microscopy experiments. Nevertheless, cell death is difficult to detect without perturbing experiments with stains, dyes or biosensors that can bias experimental outcomes, lead to inconsistent results, and reduce the number of processes that can be simultaneously labelled. These additional steps also make live microscopy difficult to scale for high-throughput screening because of the cost, labor, and analysis they entail. We address this fundamental limitation of live microscopy with biomarker-optimized convolutional neural networks (BO-CNN): computer vision models trained with a ground truth biosensor that detect live cells with superhuman, 96% accuracy more than 100 times faster than previous methods. Our models learn to identify important morphological characteristics associated with cell vitality without human input or additional perturbations, and to generalize to other imaging modalities and cell types for which they have no specialized training. We demonstrate that we can interpret decisions from BO-CNN models to gain biological insight into the patterns they use to achieve superhuman accuracy. The BO-CNN approach is broadly useful for live microscopy, and affords a powerful new paradigm for advancing the state of high-throughput imaging in a variety of contexts.


Circulation ◽  
2020 ◽  
Vol 142 (Suppl_3) ◽  
Author(s):  
Connor G OBrien ◽  
Evgeniya Vaskova ◽  
Yuko Tada ◽  
Jihye Jung ◽  
Gentaro Ikeda ◽  
...  

Introduction: Coronary artery disease is a leading cause of death worldwide. Ischemic injury leads to myocardial dysfunction, resulting in heart failure. Exosomes have emerged as a promising therapeutic for restoring the failing heart. Fundamental questions such as cell of origin and molecular cargo for optimal therapeutic effect are areas of intense research. Our lab has shown that the exosomes from bone marrow derived mesenchymal stem cells (MSC-Ex) and induced pluripotent stem cell derived cardiomyocytes (iCM-Ex) both restore injured murine myocardium. These results led us to compare the therapeutic effects of MSC-Ex vs. iCM-Ex in a porcine myocardial ischemia reperfusion (IR) injury model, a step toward predicting efficacy in humans. Hypothesis: iCM-Ex is superior to MSC-Ex in restoring the injured porcine myocardium. Methods and Results: Pigs underwent ischemia reperfusion (IR) injury, consisting of 1 hour percutaneous balloon occlusion of the proximal left anterior descending artery immediately distal to the first septal artery. Following IR injury, 5 x 10 11 exosomes were delivered in ten, 500μL intramyocardial injections using a BioCardio Helix™ catheter. Biplane ventriculography was used to target the peri-infarct region. At 2- and 4-weeks post-infarct, pigs underwent cardiac MRI (cMRI) with ciné, delayed-enhanced (DEMRI) and manganese-enhanced (MEMRI) MRI. Pigs treated with iCM-Ex (n = 5) demonstrated a 41% improvement in left ventricular ejection fraction (LVEF, p = 0.004) and 35% reduction in indexed left ventricular end diastolic volume (p = 0.008) compared to controls while MSC-Ex (N = 5) did not demonstrate significant functional improvement. Furthermore, DEMRI and MEMRI showed a 21% reduction in myocardial scar (p = 0.14) in iCM-Ex treated animals compared to control while MSC-Ex group showed no difference. RNA-seq of the exosomes and transcriptomic analysis of the ex vivo myocardium will delineate the molecular mechanism of action and the putative intracellular pathway. Conclusion: iCM-Ex is superior to MSC-Ex in improving LVEF and reducing myocardial scar formation following ischemic insult. Comparative analysis between iCM-Ex and MSC-Ex is underway to identify the molecular targets that restore the failing heart.


Author(s):  
Hisham F. Bahmada ◽  
Mohamad K. Elajami ◽  
Reem Daouk ◽  
Hiba Jalloul ◽  
Batoul Darwish ◽  
...  

: Stem cells are undifferentiated cells with the ability to proliferate and convert to different types of differentiated cells that make up the various tissues and organs in the body. They exist both in embryos as pluripotent stem cells that can differentiate into the three germ layers and as multipotent or unipotent stem cells in adult tissues to aid in repair and homeostasis. Perturbations in these cells’ normal functions can give rise to a wide variety of diseases. In this review, we discuss the origin of different stem cell types, their properties and characteristics, their role in tissue homeostasis, current research, and their potential applications in various life-threatening diseases. We focus on neural stem cells, their role in neurogenesis and how they can be exploited to treat diseases of the brain including neurodegenerative diseases and cancer. Next, we explore current research in induced pluripotent stem cell (iPSC) techniques and their clinical applications in regenerative and personalized medicine. Lastly, we tackle a special type of stem cells called cancer stem cells (CSCs) and how they can be responsible for therapy resistance and tumor recurrence and explore ways to target them.


2018 ◽  
Author(s):  
Aleksandra Klimas ◽  
Gloria Ortiz ◽  
Steven Boggess ◽  
Evan W. Miller ◽  
Emilia Entcheva

AbstractCombined optogenetic stimulation and optical imaging permits scalable, high-throughput probing of cellular electrophysiology and optimization of stem-cell derived excitable cells, such as neurons and muscle cells. We report a new “on-axis” configuration of OptoDyCE, our all-optical platform for studying human induced pluripotent stem-cell-derived cardiomyocytes (hiPSC-CMs) and other cell types, optically driven by Channelrhodopsin2 (ChR2). This solid-state system integrates optogenetic stimulation with temporally-multiplexed simultaneous recording of membrane voltage (Vm) and intracellular calcium ([Ca2+]i) dynamics using a single photodetector. We demonstrate the capacity for combining multiple spectrally-compatible actuators and sensors, including newer high-performance near-infrared (NIR) voltage probes BeRST1 and Di-4-ANBDQBS, to record complex spatiotemporal responses of hiPSC-CMs to drugs in a high-throughput manner.


Sign in / Sign up

Export Citation Format

Share Document