Network


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

Hotspot


Dive into the research topics where Armon Sharei is active.

Publication


Featured researches published by Armon Sharei.


Proceedings of the National Academy of Sciences of the United States of America | 2013

A vector-free microfluidic platform for intracellular delivery.

Armon Sharei; Janet Zoldan; Andrea Adamo; Woo Young Sim; Nahyun Cho; Emily L. Jackson; Shirley Mao; Sabine Schneider; Min-Joon Han; Abigail K. R. Lytton-Jean; Pamela Basto; Siddharth Jhunjhunwala; Jungmin Lee; Daniel A. Heller; Jeon Woong Kang; George C. Hartoularos; Kwang-Soo Kim; Daniel G. Anderson; Robert Langer; Klavs F. Jensen

Intracellular delivery of macromolecules is a challenge in research and therapeutic applications. Existing vector-based and physical methods have limitations, including their reliance on exogenous materials or electrical fields, which can lead to toxicity or off-target effects. We describe a microfluidic approach to delivery in which cells are mechanically deformed as they pass through a constriction 30–80% smaller than the cell diameter. The resulting controlled application of compression and shear forces results in the formation of transient holes that enable the diffusion of material from the surrounding buffer into the cytosol. The method has demonstrated the ability to deliver a range of material, such as carbon nanotubes, proteins, and siRNA, to 11 cell types, including embryonic stem cells and immune cells. When used for the delivery of transcription factors, the microfluidic devices produced a 10-fold improvement in colony formation relative to electroporation and cell-penetrating peptides. Indeed, its ability to deliver structurally diverse materials and its applicability to difficult-to-transfect primary cells indicate that this method could potentially enable many research and clinical applications.


Nature | 2016

In vitro and ex vivo strategies for intracellular delivery

Martin P. Stewart; Armon Sharei; Xiaoyun Ding; Gaurav Sahay; Robert Langer; Klavs F. Jensen

Intracellular delivery of materials has become a critical component of genome-editing approaches, ex vivo cell-based therapies, and a diversity of fundamental research applications. Limitations of current technologies motivate development of next-generation systems that can deliver a broad variety of cargo to diverse cell types. Here we review in vitro and ex vivo intracellular delivery approaches with a focus on mechanisms, challenges and opportunities. In particular, we emphasize membrane-disruption-based delivery methods and the transformative role of nanotechnology, microfluidics and laboratory-on-chip technology in advancing the field.


Nano Letters | 2012

Nonendocytic delivery of functional engineered nanoparticles into the cytoplasm of live cells using a novel, high-throughput microfluidic device.

Jungmin Lee; Armon Sharei; Woo Young Sim; Andrea Adamo; Robert Langer; Klavs F. Jensen; Moungi G. Bawendi

The ability to straightforwardly deliver engineered nanoparticles into the cell cytosol with high viability will vastly expand the range of biological applications. Nanoparticles could potentially be used as delivery vehicles or as fluorescent sensors to probe the cell. In particular, quantum dots (QDs) may be used to illuminate cytosolic proteins for long-term microscopy studies. Whereas recent advances have been successful in specifically labeling proteins with QDs on the cell membrane, cytosolic delivery of QDs into live cells has remained challenging. In this report, we demonstrate high throughput delivery of QDs into live cell cytoplasm using an uncomplicated microfluidic device while maintaining cell viabilities of 80-90%. We verify that the nanoparticle surface interacts with the cytosolic environment and that the QDs remain nonaggregated so that single QDs can be observed.


Nature Communications | 2016

Live-cell protein labelling with nanometre precision by cell squeezing

Alina Kollmannsperger; Armon Sharei; Anika Raulf; Mike Heilemann; Robert Langer; Klavs F. Jensen; Ralph Wieneke; Robert Tampé

Live-cell labelling techniques to visualize proteins with minimal disturbance are important; however, the currently available methods are limited in their labelling efficiency, specificity and cell permeability. We describe high-throughput protein labelling facilitated by minimalistic probes delivered to mammalian cells by microfluidic cell squeezing. High-affinity and target-specific tracing of proteins in various subcellular compartments is demonstrated, culminating in photoinduced labelling within live cells. Both the fine-tuned delivery of subnanomolar concentrations and the minimal size of the probe allow for live-cell super-resolution imaging with very low background and nanometre precision. This method is fast in probe delivery (∼1,000,000 cells per second), versatile across cell types and can be readily transferred to a multitude of proteins. Moreover, the technique succeeds in combination with well-established methods to gain multiplexed labelling and has demonstrated potential to precisely trace target proteins, in live mammalian cells, by super-resolution microscopy.


Scientific Reports | 2015

Microfluidic squeezing for intracellular antigen loading in polyclonal B-cells as cellular vaccines.

Gregory L. Szeto; Debra S. Van Egeren; Hermoon Worku; Armon Sharei; Brian Alejandro; Clara Park; Kirubel Frew; Mavis S. Brefo; Shirley Mao; Megan Heimann; Robert Langer; Klavs F. Jensen; Darrell J. Irvine

B-cells are promising candidate autologous antigen-presenting cells (APCs) to prime antigen-specific T-cells both in vitro and in vivo. However to date, a significant barrier to utilizing B-cells as APCs is their low capacity for non-specific antigen uptake compared to “professional” APCs such as dendritic cells. Here we utilize a microfluidic device that employs many parallel channels to pass single cells through narrow constrictions in high throughput. This microscale “cell squeezing” process creates transient pores in the plasma membrane, enabling intracellular delivery of whole proteins from the surrounding medium into B-cells via mechano-poration. We demonstrate that both resting and activated B-cells process and present antigens delivered via mechano-poration exclusively to antigen-specific CD8+T-cells, and not CD4+T-cells. Squeezed B-cells primed and expanded large numbers of effector CD8+T-cells in vitro that produced effector cytokines critical to cytolytic function, including granzyme B and interferon-γ. Finally, antigen-loaded B-cells were also able to prime antigen-specific CD8+T-cells in vivo when adoptively transferred into mice. Altogether, these data demonstrate crucial proof-of-concept for mechano-poration as an enabling technology for B-cell antigen loading, priming of antigen-specific CD8+T-cells, and decoupling of antigen uptake from B-cell activation.


Analytical Chemistry | 2013

Flow-through comb electroporation device for delivery of macromolecules.

Andrea Adamo; Alessandro Arione; Armon Sharei; Klavs F. Jensen

We present a microfluidic electroporation device with a comb electrode layout fabricated in polydimethylsiloxane (PMDS) and glass. Characterization experiments with HeLa cells and fluorescent dextran show efficient delivery (∼95%) with low toxicity (cell viability ∼85%) as well as rapid pore closure after electroporation. The activity of delivered molecules is also verified by silencing RNA (siRNA) studies that demonstrate gene knockdown in GFP expressing cells. This simple, scalable approach to microfluidic, flow-through electroporation could facilitate the integration of electroporation modules within cell analysis devices that perform multiple operations.


Nature Biomedical Engineering | 2017

High-throughput nuclear delivery and rapid expression of DNA via mechanical and electrical cell-membrane disruption

Xiaoyun Ding; Martin P. Stewart; Armon Sharei; James C. Weaver; Robert Langer; Klavs F. Jensen

Nuclear transfection of DNA into mammalian cells is challenging yet critical for many biological and medical studies. Here, by combining cell squeezing and electric-field-driven transport in a device that integrates microfluidic channels with constrictions and microelectrodes, we demonstrate nuclear delivery of plasmid DNA within 1 hour after treatment, the most rapid DNA expression in a high-throughput setting (up to millions of cells per minute per device). Passing cells at high speed through microfluidic constrictions smaller than the cell diameter mechanically disrupts the cell membrane, allowing a subsequent electric field to further disrupt the nuclear envelope and drive DNA molecules into the cytoplasm and nucleus. By tracking the localization of the ESCRT-III (endosomal sorting complexes required for transport) protein CHMP4B, we show that the integrity of the nuclear envelope is recovered within 15 minutes of treatment. We also provide insight into subcellular delivery by comparing the performance of the disruption-and-field-enhanced method with those of conventional chemical, electroporation, and manual-injection systems.


PLOS ONE | 2015

Ex Vivo Cytosolic Delivery of Functional Macromolecules to Immune Cells

Armon Sharei; Radiana Trifonova; Siddharth Jhunjhunwala; George C. Hartoularos; Alexandra T. Eyerman; Abigail K. R. Lytton-Jean; Mathieu Angin; Siddhartha Sharma; Roberta Poceviciute; Shirley Mao; Megan Heimann; Sophia Liu; Tanya Talkar; Omar F. Khan; Marylyn M. Addo; Ulrich H. von Andrian; Daniel G. Anderson; Robert Langer; Judy Lieberman; Klavs F. Jensen

Intracellular delivery of biomolecules, such as proteins and siRNAs, into primary immune cells, especially resting lymphocytes, is a challenge. Here we describe the design and testing of microfluidic intracellular delivery systems that cause temporary membrane disruption by rapid mechanical deformation of human and mouse immune cells. Dextran, antibody and siRNA delivery performance is measured in multiple immune cell types and the approach’s potential to engineer cell function is demonstrated in HIV infection studies.


Journal of Visualized Experiments | 2013

Cell Squeezing as a Robust, Microfluidic Intracellular Delivery Platform

Armon Sharei; Nahyun Cho; Shirley Mao; Emily L. Jackson; Roberta Poceviciute; Andrea Adamo; Janet Zoldan; Robert Langer; Klavs F. Jensen

Rapid mechanical deformation of cells has emerged as a promising, vector-free method for intracellular delivery of macromolecules and nanomaterials. This technology has shown potential in addressing previously challenging applications; including, delivery to primary immune cells, cell reprogramming, carbon nanotube, and quantum dot delivery. This vector-free microfluidic platform relies on mechanical disruption of the cell membrane to facilitate cytosolic delivery of the target material. Herein, we describe the detailed method of use for these microfluidic devices including, device assembly, cell preparation, and system operation. This delivery approach requires a brief optimization of device type and operating conditions for previously unreported applications. The provided instructions are generalizable to most cell types and delivery materials as this system does not require specialized buffers or chemical modification/conjugation steps. This work also provides recommendations on how to improve device performance and trouble-shoot potential issues related to clogging, low delivery efficiencies, and cell viability.


ACS Chemical Biology | 2017

Microfluidic-Enabled Intracellular Delivery of Membrane Impermeable Inhibitors to Study Target Engagement in Human Primary Cells

Jing Li; Bu Wang; Brian Juba; Michael L. Vazquez; Steve W. Kortum; Betsy S. Pierce; Michael Pacheco; Lee R. Roberts; Joseph Walter Strohbach; Lyn H. Jones; Erik C. Hett; Atli Thorarensen; Jean-Baptiste Telliez; Armon Sharei; Mark Edward Bunnage; Jonathan B. Gilbert

Biochemical screening is a major source of lead generation for novel targets. However, during the process of small molecule lead optimization, compounds with excellent biochemical activity may show poor cellular potency, making structure-activity relationships difficult to decipher. This may be due to low membrane permeability of the molecule, resulting in insufficient intracellular drug concentration. The Cell Squeeze platform increases permeability regardless of compound structure by mechanically disrupting the membrane, which can overcome permeability limitations and bridge the gap between biochemical and cellular studies. In this study, we show that poorly permeable Janus kinase (JAK) inhibitors are delivered into primary cells using Cell Squeeze, inhibiting up to 90% of the JAK pathway, while incubation of JAK inhibitors with or without electroporation had no significant effect. We believe this robust intracellular delivery approach could enable more effective lead optimization and deepen our understanding of target engagement by small molecules and functional probes.

Collaboration


Dive into the Armon Sharei's collaboration.

Top Co-Authors

Avatar

Klavs F. Jensen

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Robert Langer

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Andrea Adamo

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Shirley Mao

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Nahyun Cho

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Roberta Poceviciute

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Camilo Ruiz

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Darrell J. Irvine

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Emily L. Jackson

Massachusetts Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge