Happening @ Michigan https://events.umich.edu/list/rss RSS Feed for Happening @ Michigan Events at the University of Michigan. BME PhD Defense: Feiran Li (June 13, 2022 12:00pm) https://events.umich.edu/event/95461 95461-21789961@events.umich.edu Event Begins: Monday, June 13, 2022 12:00pm
Location: North Campus Research Complex Building 520
Organized By: Biomedical Engineering

Cell-based therapies are emerging for Type I diabetes mellitus (T1D), an autoimmune disease characterized by the destruction of insulin producing pancreatic β-cells, as a means to provide long term restoration of glycemic control. The limited supply of donor islets has motivated research into methods for differentiating pancreatic β-cells from renewable pluripotent stem cells such as human pluripotent stem cells (hPSCs). Biomaterial scaffolds maintain the integrity of cell-to-cell and cell-to-matrix connections by avoiding the disruption of the cell niche during handling. This dissertation addresses three key questions with respect to cell therapy and immunomodulation for T1D, including culture system on porous PLG scaffold, functionalized scaffold for improved cell viability and maturation, and immunomodulation with the membrane coated nanoparticles (MCNPs).

Culture on porous biomaterial scaffolds of hPSCs was investigated at multiple stages of differentiation between Stage 0 and 6 for improved differentiation. Scaffolds are biomaterial devices that could provide chemical and physical cues to control the microenvironment and subsequently alter cellular behavior by facilitating cell-cell interactions. The culture of cells on the scaffolds was found to support maturation of SC derived beta cells depending on the stage of seeding. Suspension cultured-pancreatic progenitors seeded onto scaffolds for stage 5 culture (pancreatic endocrine development), demonstrated enhanced expression for many maturation genes compared to cells that remained in suspension culture through the end of stage 6. This study showcased the scaffold culture as a promising platform for maturation that allows cells to develop a niche and may allow for direct transplantation without manipulating cells.

Early engraftment and development of β-cells post transplantation are a major limitation for stem cell derived beta cells due in part to their being immature. The survival and development of hPSC-derived β-cells seeded onto PLG microporous scaffolds were investigated within the initial 2 weeks post transplantation. Early inflammatory events induced by the biomaterial and transplanted cells heavily affected hPSC-derived β-cell engraftment due to the innate immune response. The inflammation includes the production of soluble mediators, inflammatory cytokines and the recruitment of innate cells at the graft site, hindering early graft engraftment and in-vivo hPSC-derived β-cell maturation. The PLG-based biodegradable scaffold chemically linked with a novel form of FasL chimeric with streptavidin, SA-FasL, was applied to create an immunoprivileged transplant site by modulating the local inflammatory microenvironment. The β-cell viability and differentiation were found improved at the SA-FasL induced immunoprivileged site together with a suppressed inflammatory reaction.

Life-long systemic immune suppression due to allogenic graft/cell transplant also limits the translation of cell therapies for T1D. We investigated the design of membrane-coated nanoparticles (MCNPs), with membranes derived from bone marrow-derived dendritic cells and coated onto poly(lactic-co-glycolic acid) (PLGA) nanoparticle cores, to directly interact with both naïve and activated T cells. Mechanistic studies revealed that the developed MCNPs have the capability to communicate with allogenic T cells by modulating the cytokine secretion levels similar to professional antigen presenting cells. Furthermore, the MCNPs can be engineered pre- and post-fabrication for upregulated surface molecules or varied antigen binding and can be functionalized by biotinylation for a wider range of protein loading.

Overall, this dissertation discussed optimization and early immunomodulation of the biomaterial culturing system for hPSC-derived β cells, and development of tunable MCNPs for direct T cell communication.

Date: Monday, June 13, 2022
Time: 12:00 PM
Location: NCRC Building 520 Room 1122 and Zoom (https://umich.zoom.us/j/93840656651)
Chair: Dr. Lonnie Shea

]]>
Lecture / Discussion Tue, 07 Jun 2022 09:22:42 -0400 2022-06-13T12:00:00-04:00 2022-06-13T13:00:00-04:00 North Campus Research Complex Building 520 Biomedical Engineering Lecture / Discussion BME Defense
Understanding and engineering microbes for solving complex problems in biology and medicine (June 16, 2022 3:30pm) https://events.umich.edu/event/95526 95526-21790074@events.umich.edu Event Begins: Thursday, June 16, 2022 3:30pm
Location: Lurie Biomedical Engineering (formerly ATL)
Organized By: Biomedical Engineering

Abstract:

The microbiome represents an exciting frontier in medicine, and early successes in the field have demonstrated the dynamic interactions among individual microbial species and highlighted the crosstalk between microbiota and their hosts at the mucosal interface.  The Li research group in the Department of Bioengineering at Northeastern University focuses on the development of molecular and live cell-based therapeutics, with a major emphasis on harnessing innovative synthetic biology and drug delivery approaches for improving human health in a sustainable manner. In this talk, I will present our work from the past three years in interrogating and manipulating commensal bacteria and probiotics as therapeutic platforms to promote human health.

Bio:

Jiahe Li obtained his PhD in Biomedical Engineering at Cornell University in 2015, where he leveraged synthetic biology approaches and cell biology to engineer bacteria and platelets as platforms for treating metastatic cancer. Later, he pursued his postdoctoral training at the Koch Institute for Integrative Cancer Research at MIT from 2015-2018, where he gained complementary expertise in polymer science and gene delivery. He started a tenure-track faculty position in the Department of Bioengineering at Northeastern University in 2019, and his current research is supported by NIH, DoD, and various biotech companies.

Zoom Link: https://umich.zoom.us/j/97247012805

]]>
Lecture / Discussion Thu, 09 Jun 2022 15:34:33 -0400 2022-06-16T15:30:00-04:00 2022-06-16T16:30:00-04:00 Lurie Biomedical Engineering (formerly ATL) Biomedical Engineering Lecture / Discussion BME Seminar
Discovery and Development of Agonist Antibodies for T Cell Receptors (July 29, 2022 10:00am) https://events.umich.edu/event/96254 96254-21792188@events.umich.edu Event Begins: Friday, July 29, 2022 10:00am
Location: Off Campus Location
Organized By: Biomedical Engineering

Agonist antibodies that activate co-stimulatory immune receptors, such as the tumor necrosis factor (TNF) receptors OX40 and CD137, are an important class of emerging therapeutics due to their ability to regulate immune cell activity. Despite their promise, there are no approved agonist antibodies for treating cancer as demonstrated by previous unsuccessful clinical trials. Although multiple factors are responsible for poor clinical efficacy, one major bottleneck is the reliance on FcγR-mediated crosslinking for sufficient receptor activation. This is inherently problematic because FcγR expression varies greatly on different immune cells, leading to a wide range of receptor agonism. Emerging research suggests that antibodies engaging two different epitopes on the same immune receptor mediate receptor superclustering and enable robust antibody agonism without extrinsic Fc crosslinking. However, there are no systematic methods for identifying such biepitopic (also known as biparatopic) agonist antibodies. Therefore, the objective of this research work is to develop facile methods for reliably identifying biepitopic antibodies to activate immune receptors for immunotherapeutic applications.

Biepitopic antibodies have been shown to mediate potent receptor activation for a variety of immune receptors. Traditionally, the generation of these antibodies requires key steps including animal immunization, epitope binning to identify unique antibody pairs, and combining antibody pairs to engineer biepitopic antibodies. While this approach has been used to successfully discover biepitopic antibodies, it suffers from key limitations. Notably, animal immunization and subsequent antibody isolation is an arduous and unpredictable process. Even when successful clones are discovered from these processes, further epitope binning experiments are needed to select antibody pairs to discover potent immune therapeutics. To overcome these limitations, we developed an antibody screening strategy that greatly simplifies the discovery of biepitopic antibodies. Our approach eliminates the need for animal immunization by using existing, off-the-shelf IgG antibodies specific to the target receptor. Next, we perform in vitro selections by blocking the receptor epitope of the existing antibody and conducting subsequent sorts to identify single-chain antibodies with orthogonal binding domains. Thus far, our work has shown that the antibody screening strategy can be used to discover antibodies for a variety of TNF receptors including OX40 and CD137.

Given that receptor clustering of three or more receptors is critical for activating TNF receptors, we first generated biepitopic tetravalent OX40 antibodies by attaching novel single-chain antibodies to the C-termini of the light chain of existing clinical-stage antibodies. These tetravalent biepitopic antibodies showed remarkable T cell proliferation and cytokine secretion for biepitopic antibodies compared to their monoepitopic counterparts. Next, we sought to improve the additional clinical-stage OX40 IgGs engineered as biepitopic antibodies to show the generality of our findings that biepitopic antibodies can mediate superior and FcγR-independent activities. Beyond OX40 IgGs, we also show that biepitopic antibodies can be used to mediate superior T cell proliferation for other TNF receptors including CD137. Looking forward, we anticipate that these research advancements will accelerate the discovery and development of the next generation of immune therapeutics.

Date: Friday, July 29, 2022
Time: 10:00 AM
Zoom: https://umich.zoom.us/j/5163583658
Co-Chairs: Professors Peter Tessier and Lonnie Shea

]]>
Lecture / Discussion Tue, 26 Jul 2022 13:09:39 -0400 2022-07-29T10:00:00-04:00 2022-07-29T11:00:00-04:00 Off Campus Location Biomedical Engineering Lecture / Discussion BME Ph.D. Defense
Hierarchical motion modeling of abdominal motions for radiation therapy (August 9, 2022 10:00am) https://events.umich.edu/event/96536 96536-21792631@events.umich.edu Event Begins: Tuesday, August 9, 2022 10:00am
Location: Lurie Biomedical Engineering (formerly ATL)
Organized By: Biomedical Engineering

Abstract:

Human abdominal organs are subject to a variety of physiological forces that superimpose their effects to influence local motion and configuration. Motions include breathing, gastric contraction, and other types of less periodic slow configuration changes. Breathing motion has been extensively studied and well characterized; however, gastric contraction and slow configuration motion have been rarely investigated. By using a golden angle stack-of-star radial sampling magnetic resonance image (MRI) sequence, we constructed a hierarchical motion model that characterizes each of these three motions, as well as their combined effects. Breathing motion is extracted and corrected as the first step, following by reconstruction of gastric motion and slow configuration changes. The model shows non-neglectable geometric displacements raised by all three motion modes. These motions, if not managed properly during radiation therapy, may potentially result in overdose to normal tissue or underdosage to the tumor target. Magnetic resonance guided radiotherapy (MRgRT) systems have been developed which have the technical capability to address these complex motions, but to date their primary applications have been relegated to management of breathing motion. In this dissertation, we proposed a gastric motion prediction framework to allow real-time management of contractile motion during MRgRT taking advantage of the intra-scan stability of gastric contraction motion observed in patients under standard pre-session eating restrictions. The framework was able to achieve submillimeter prediction error with a sufficient future prediction time to overcome the latency introduced by the image sampling reconstruction, motion assessment and treatment interruption or modification on MR-guided linear accelerators. Motions and deformations during radiation treatment present a challenge to precisely and accurately measure the radiation dose delivered to abdominal organs. A dose accumulation tool, developed based on the hierarchical motion model, was built to estimate dose distributions with abdominal motions. The tool demonstrates potential deviations of dose due to motion and shows exceeding of dose constraints in certain cases. It could support offline adaptation or help record delivered dose more accurately than stationary images used for daily patient positioning and/or online adaptation of treatment plans. The motion model is also currently supporting other clinical applications, including providing improved image quality reconstructions from free-breathing scans for improvement of accuracy of perfusion as well as liver functional maps. In the future, the model can be further utilized in other fields including radiology or gastroenterology.  

Room: LBME 2185 / Zoom Link: https://umich.zoom.us/j/93620134849 Meeting ID: 936 2013 4849 Passcode: 268890

Committee Chair(s): Dr. James Balter and Dr. Rojano Kashani

]]>
Lecture / Discussion Fri, 05 Aug 2022 10:05:16 -0400 2022-08-09T10:00:00-04:00 2022-08-09T11:00:00-04:00 Lurie Biomedical Engineering (formerly ATL) Biomedical Engineering Lecture / Discussion BME Defense
Effects of Electric Stimulation on Physiology and Anatomy of the Visual Pathway (August 10, 2022 12:00pm) https://events.umich.edu/event/96539 96539-21792637@events.umich.edu Event Begins: Wednesday, August 10, 2022 12:00pm
Location:
Organized By: Biomedical Engineering

Abstract:
Retinal degenerative diseases that progressively lead to severe blindness impact the affected individual’s quality-of-life. Visual prosthesis technology aims to provide an individual a potential means of obtaining visual information lost to them by blindness. Since the proof-of-concept success in 1968 of a device implanted in a human, visual prostheses have had sustained academic research and commercial interest. However, commercial failure of two retinal prosthesis device has raised concerns for the visual prosthesis field. To learn from this experience, research in this dissertation is aimed at understanding the impact of electric stimulation on the target neural tissue and investigating technology for a visual cortex prosthesis, which can reach a larger patient population (compared to a retinal prosthesis).

My first set of experiments assessed, in an animal model of retinal degeneration, the condition of the brain and its ability to receive artificial vision information. Retinitis Pigmentosa has been proven to impact the human brain. My study investigated the extent to which this was replicated in a rat animal model of a single genetic mutation of Retinitis Pigmentosa. The P23H-1 rat was investigated with electrophysiology and immunohistochemistry to understand the brain’s function and structural condition. The rat brain’s response to light and electric stimulation was investigated, and the change of visually evoked responses and maintenance of electrically evoked responses was observed. Histology images show a relatively stable macrostructure of the blind rat brain.

I also performed retinal and cortical implant procedures to test newly developed visual prosthesis technology to enable investigations into researching neural change occurring from blindness and electric stimulation. A retinal device with Parylene-C as its main component was tested and its feasibility in the small eye of a rat animal model was investigated. The device can survive 4-weeks of implantation and is stable within the eye. In support of the development of a novel cortical visual prosthesis device that fits the need of blind individuals, I used a small animal model first to prove the efficacy and safety of a novel neurostimulation electrode. The device, named StiMote, is in preclinical development. I worked to characterize the full ability of the neural interface, High-Density Carbon Fibers with electrodeposited Platinum-Iridium. The ability of PtIr-HDCF as a recording and stimulation neural interface device was verified using electrochemical measurements before, during, and after a long-duration 7-hour electric stimulation session that simulates a full day of device use.

PtIr-HDCF as a neural interface device was verified by my previous work and its improvement in reducing neuroinflammatory response compared to other microelectrode array archetypes has been previously researched. As a result, PtIr-HDCF can be used as a device to monitor the brain and can better extract the effect of electric stimulation on the brain alone. I recorded neural electrophysiology to verify the rat brain’s sensitivity to stimuli before and after 7-hour stimulation. To supplement the already existing neural implant and electric stimulation inflammation data, Spatial Transcriptomics as a novel method to define electric stimulation safety was performed. Spatial Transcriptomics showed that PtIr-HDCF, when compared to a conventional microwire array, performs better in sustaining neural health by reducing neuroinflammation and eliciting mRNA upregulation of neurotrophic factors.

Findings of this project can be used to better inform future investigations into brain electrophysiology and transcriptomics projects aimed to understand the neural change from blindness and electric stimulation.

Committee Chair(s): Dr. James Weiland

Location: 1501 Auditorium, NCRC Bldg 32 & https://umich.zoom.us/j/91500987159?pwd=RWIvQkZVT2FHZjQ2S1BBS2k0ck1SUT09

]]>
Lecture / Discussion Fri, 05 Aug 2022 10:32:21 -0400 2022-08-10T12:00:00-04:00 2022-08-10T13:00:00-04:00 Biomedical Engineering Lecture / Discussion BME Defense
Data-driven Methods for Automated Assessment of Coronary Artery Disease (August 30, 2022 9:00am) https://events.umich.edu/event/97256 97256-21794237@events.umich.edu Event Begins: Tuesday, August 30, 2022 9:00am
Location: Lurie Robert H. Engin. Ctr
Organized By: Biomedical Engineering

Abstract:

The current gold standard for Coronary Artery Disease (CAD) diagnosis is X-ray angiography. Visual estimation can be subjective, therefore semi-automated software tools such as Quantitative Coronary Angiography (QCA) have been developed to quantify disease severity. Alternatively, functional metrics such as Fractional Flow Reserve (FFR) have demonstrated better diagnostic outcomes than anatomical assessment, but they are not widely used due to cost and risk. Ideally, quantitative and functional information could be derived directly from X-ray angiography images without the additional risks, time, and cost associated with performing FFR or QCA.

The goal of this project is to develop automated data-driven approaches for anatomical and functional quantification of disease severity using X-ray angiography images. To this end, we have developed algorithms for 1) automated coronary vessel segmentation, 2) stenosis detection and characterization, 3) 3D reconstruction of coronary anatomy, and 4) image-based flow extraction. These algorithms can be used in conjunction with computational fluid dynamics (CFD) modeling to assess the functional significance of disease.

We first present AngioNet, a neural network for coronary segmentation from X-ray angiography images. Conventional algorithms relying on thresholding or filtering cannot distinguish between the coronary vessels and the catheter used to inject the dye. AngioNet’s key innovation is an Angiographic Processing Network, or APN, which learns the best possible combination of pre-processing filters to improve segmentation performance. AngioNet demonstrates state-of-the-art segmentation accuracy (Dice score = 0.864) and does not segment the catheter in challenging cases where other neural networks fail.

Building upon AngioNet, we developed combination of neural networks and image processing algorithms to automatically localize, segment, and measure stenoses. This pipeline was able to measure stenosis diameter within 0.206±0.155mm or approximately 1 pixel of ground truth measurements from QCA. It is also the first automated pipeline to quantify rather than categorize disease severity.

Although measuring stenosis diameter in 2D images is useful, a more robust approach would be to measure diameters in the 3D coronary anatomy. Another advantage of the 3D coronary anatomy is that it can be used to perform CFD simulations of blood flow and compute functional metrics such as FFR. To this end, we developed a machine learning approach for automated 3D vessel reconstruction from a series of uncalibrated 2D X-ray angiography images. This approach is superior to projective geometry methods for 3D reconstruction due to their semi-automatic nature and reliance on accurate knowledge of input image acquisition angles. Our machine learning approach has demonstrated sub-pixel error in radius reconstruction (0.16±0.07mm) and 1% error in FFR computed in a reconstructed coronary tree.

In addition to the 3D coronary geometry, information about patient-specific flow or pressure is required to perform a hemodynamics simulation and compute FFR. We developed an algorithm that tracks vessel area in sequential frames of a segmented angiography series to estimate relative flow in each branch. We validated the algorithm in the simplest possible case, using a simulation of dye transport under steady flow conditions as the ground truth. On average, the difference in relative flow per branch was 5.15% for a healthy coronary tree and 3.68% in a coronary tree with stenosis.

We finally demonstrated the successes and limitations of the methods developed in this thesis by comparing computational FFR derived using the above algorithms against clinically measured FFR. The error between the calculated and clinically measured FFR was 0.1, corresponding to an 11% error.

Committee Chair(s):
Dr. C. Alberto Figueroa and Dr. Brahmajee K. Nallamothu

Zoom Link: https://umich.zoom.us/j/94364657250, Passcode: 390041 *Registration is required

]]>
Workshop / Seminar Mon, 22 Aug 2022 16:54:04 -0400 2022-08-30T09:00:00-04:00 2022-08-30T10:00:00-04:00 Lurie Robert H. Engin. Ctr Biomedical Engineering Workshop / Seminar BME Defense Announcement
A Novel Bioelastomer Platform with Tailorable Design Parameters for Cartilage Regeneration (August 30, 2022 11:00am) https://events.umich.edu/event/97426 97426-21794553@events.umich.edu Event Begins: Tuesday, August 30, 2022 11:00am
Location: Lurie Biomedical Engineering (formerly ATL)
Organized By: Biomedical Engineering

Abstract:

Articular cartilage has limited ability to self-repair, which often causes focal defects to progress into post traumatic osteoarthritis. Autologous chondrocyte implantation, a process in which chondrocytes are harvested from the patient, expanded in monolayer culture, and injected into the defect site, is one of the most common approaches to treat cartilage defect. However, chondrocyte dedifferentiation during this process reduces their ability to durably restore cartilage function. Chondrocyte-based cartilage tissue engineering offers alternative approaches for cartilage repair to overcome the limitations of current clinical options by developing environments that combines cues from synthetic scaffold and biological factors to enhance chondrocyte function. However, the translation to the clinic has been limited by our incomplete understanding of how scaffold design parameters interact together to control cell function. Therefore, this dissertation focuses on designing a chondrocyte-based biomaterial platform made with a novel elastomeric synthetic scaffold, poly(glycerol dodecanedioate) (PGD), to investigate the combinatory effects of design parameters on chondrocytes behavior in vitro.

First, this thesis evaluates the effects of surface modification of PGD on the shape and extracellular matrix (ECM) production of chondrocyte, both of which are crucial for robust cartilage formation. I investigated two different strategies to generate a biomaterial surface with high cell affinity: 1) coating with various concentration of collagen type I or hyaluronic acid individually or in combination, or 2) altering the surface charge and roughness using various level of alkaline hydrolysis. Our results revealed the combinatorial effects of ligand composition and density or surface charge and roughness on human articular chondrocyte function.

Lastly, I used finite element analysis to determine if the local strain fields that developed inside the pores under load could be tuned to be within the range shown to have an anabolic effect on chondrocyte function. The tensile strains that develop along 31% – 71% pore surfaces inside of porous PGD scaffolds, according to varying pore size and porosity, were at levels shown to stimulate chondrocyte ECM production, indicating that the pore structural parameters could be tuned to optimize cellular-level strain profiles. These results suggest that porous PGD scaffolds have the potential to guide cartilage regeneration.

Overall, this dissertation produces a platform for cartilage tissue engineering using a novel bioelastomer PGD, in which the scaffold design parameters, such as surface modification and cellular strain, can be modified to enhance chondrocyte function.

Committee Chair(s):
Dr. Rhima Coleman

Zoom Link: https://umich.zoom.us/j/91391389305, Passcode: PGD

]]>
Presentation Thu, 25 Aug 2022 11:41:37 -0400 2022-08-30T11:00:00-04:00 2022-08-30T12:00:00-04:00 Lurie Biomedical Engineering (formerly ATL) Biomedical Engineering Presentation BME Ph.D. Defence
From molecules to development: biological timing and patterning (September 8, 2022 4:30pm) https://events.umich.edu/event/97913 97913-21795312@events.umich.edu Event Begins: Thursday, September 8, 2022 4:30pm
Location: Electrical Engineering and Computer Science Building
Organized By: Biomedical Engineering

Abstract:
Organisms from bacteria to humans employ complex biochemical or genetic oscillatory networks, termed biological clocks, to drive a wide variety of cellular and developmental processes for robust timing and patterning. Despite their complexity and diversity, many of these clocks share the same core architectures that are highly conserved from species to species, suggesting an essential role of network structures underlying clock functioning. The Yang lab, bridging biophysics and systems & synthetic biology, has integrated modeling with experiments in minimal cells and live embryos to elucidate universal physical mechanisms underlying the complex processes during development. In this talk, I will focus on our recent efforts in understanding the design and interaction of cellular clocks in cell cycles and embryonic developmental patterns. Computationally, we have identified network motifs, notably incoherent inputs, that universally enhance systems' robust performance. Experimentally, we developed a unique synthetic-cell system in microfluidic droplets to analyze circuits and functions of robustness and tunability. We also established single-cell assays of zebrafish embryos combined with biomechanics to analyze the role of energy and mechanical and biochemical signaling in spatiotemporal patterns.

Bio:
Qiong Yang received a Ph.D. in Physics from MIT in 2009 before joining the Department of Chemical and Systems Biology at Stanford University for postdoctoral research, supported by the Stanford Dean’s Postdoctoral Fellowship and a Damon Runyon Cancer Research Fellowship. She was appointed as an Assistant Professor in Biophysics at the University of Michigan in 2014 and was promoted to Associate Professor in 2022. Her research group is affiliated with the departments of Physics, Applied Physics, BME, Complex Systems, and Computational Medicine & Bioinformatics at UM. She has received awards including NSF CAREER, NIH MIRA, Sloan Fellowship, Elizabeth C. Crosby Award, and Class of 1923 Memorial Teaching Award.​​​​​​​

Zoom:
https://umich.zoom.us/j/91375430500

]]>
Workshop / Seminar Thu, 01 Sep 2022 10:44:42 -0400 2022-09-08T16:30:00-04:00 2022-09-08T17:30:00-04:00 Electrical Engineering and Computer Science Building Biomedical Engineering Workshop / Seminar BME 500 Seminar
Engineering Operational Transplant Tolerance via Biomaterials (September 15, 2022 4:30pm) https://events.umich.edu/event/97969 97969-21795406@events.umich.edu Event Begins: Thursday, September 15, 2022 4:30pm
Location: Electrical Engineering and Computer Science Building
Organized By: Biomedical Engineering

Abstract:
Organ and cell replacement therapies hold great promise for the treatment of multiple conditions, including autoimmune diseases such as type 1 diabetes. Restoration of endogenous insulin production, via cell delivery, has shown to be clinically successful in lowering complications and improving glucose sensing in patients. Yet, a widespread application has been hampered by the need for chronic immunosuppressive drugs to prevent strong inflammatory and immunological responses to the graft. Engineered materials offer a powerful approach for local, selective targeting of immune functionalities without compromising systemic immune function. In this talk, we will highlight engineered synthetic polymeric materials that can promote tissue integration and induce operational tolerance to cell therapies by generating a multifaced regulatory network.

Bio:
Prof Coronel is a Biological scholar and Assistant Professor of Biomedical Engineering at the University of Michigan. Her lab is centered on engineering biomaterials for perturbing and investigating immunological responses. Dr. Coronel received her BS degree in Biomedical Engineering from the University of Miami, and her Ph.D. in Biomedical Engineering from the University of Florida. She also obtained a certificate in Clinical Translational Research from Emory University Public Health School. She finished her postdoctoral fellowship at the Georgia Institute of Technology. Her work has been funded by JDRF, NIH, and the programmable materials initiative at the University of Michigan.

Zoom:
https://umich.zoom.us/j/91375430500

]]>
Workshop / Seminar Thu, 01 Sep 2022 15:04:37 -0400 2022-09-15T16:30:00-04:00 2022-09-15T17:30:00-04:00 Electrical Engineering and Computer Science Building Biomedical Engineering Workshop / Seminar BME 500 Seminar
Metabolic Reprogramming of Donor Hearts to Improve Function (September 22, 2022 4:30pm) https://events.umich.edu/event/98895 98895-21797323@events.umich.edu Event Begins: Thursday, September 22, 2022 4:30pm
Location: Electrical Engineering and Computer Science Building
Organized By: Biomedical Engineering

Abstract:
Harmful metabolic processes are well underway during cold preservation of donor hearts. We discovered a method to increase the expression of beneficial enzymes which augment the production of anti-inflammatory metabolites. This leads to lowered oxidative stress, reduced myocardial injury and translates into better cardiac function following transplantation. Future strategies to reduce primary graft dysfunction could involve precise modulation of these cardiac metabolic pathways.

Bio:
Paul Tang is an Assistant Professor of Cardiac Surgery at the University of Michigan-Ann Arbor. His cardiothoracic surgery training was completed at Duke University Medical Center where he also received advanced training in heart transplantation, ventricular assist devices and aortic surgery. He has given talks and published widely on the natural history and surgical outcomes of these diseases. At Yale University, Dr. Tang completed a PhD focused on cardiovascular immunology. Dr. Tang's clinical practice includes surgical treatment of heart failure (i.e. heart transplantation, ventricular assist devices), valvular repair or replacement, and aortic aneurysm surgery. He is an investigator in various national clinical trials for heart failure management, and is a member of professional societies such as The International Society for Heart and Lung Transplantation, Southern Thoracic Surgical Association, American Heart Association, and the Society of Thoracic Surgeons.​​​​​​​

Zoom:
https://umich.zoom.us/j/91375430500

]]>
Workshop / Seminar Fri, 16 Sep 2022 16:19:36 -0400 2022-09-22T16:30:00-04:00 2022-09-22T17:30:00-04:00 Electrical Engineering and Computer Science Building Biomedical Engineering Workshop / Seminar BME 500 Seminar
Predictive analysis and deep learning of functional MRI in Alzheimer's disease (September 26, 2022 10:00am) https://events.umich.edu/event/97916 97916-21795315@events.umich.edu Event Begins: Monday, September 26, 2022 10:00am
Location: Off Campus Location
Organized By: Biomedical Engineering

Abstract:
Alzheimer's disease (AD) and dementia pose a significant burden to individuals and public health. AD is expected to grow in prevalence in the coming decades due to the aging population. Brain atrophy is a major component of AD pathology and can occur before symptoms of cognitive impairment. However, pathological brain atrophy and symptoms of cognitive impairment may be a result of many years of disease impacts. Evidence supports the need for early detection of impacted neurocircuitry to foresee future progression to advanced stages of AD and develop treatments. This dissertation examines predictive modeling and deep learning methods to identify brain-behavior relationships and learn low-dimensional representations of brain activity from MR imaging data. The dissertation and methods are separated into four parts.  

Part one of this work examines multivariate analysis approaches applied to functional connectivity from subjects with an early clinical phenotype of AD, mild cognitive impairment (MCI). A regression framework using partial least squares and feature selection demonstrated significant brain-behavior relationships with measures of cognition and memory. The results also confirm other findings that ecologically relevant task-based connectivity serves as a ``stress-test" for memory-related deficits such as those observed in MCI. This approach elucidated brain regions that may be implicated in MCI and warrant future study (superior temporal gyrus, inferior parietal lobule, and superior frontal gyrus). Part two extends the multivariate analyses studied in part one to an additional brain imaging modality, arterial spin labeling (ASL). Cerebral blood flow (CBF) as measured by ASL demonstrated brain-behavior relationships with composite measures of memory and learning in a cohort along the spectrum of AD, demonstrating that CBF data warrant further investigation as a predictor in this application.

Parts three and four utilize a variational autoencoder (VAE) model, a deep learning approach to encode latent representations that aim to disentangle sources of fMRI signal. A surface-based VAE trained on only healthy controls is shown to be generalizable to patients with known AD pathology. The results maintained individual separation and high input/decoder output spatial reconstruction correlation of r=0.8 across all three groups. Part four extended the surface-based model used in part three to a volumetric fMRI approach. Similarly to the surface-based model, high reconstruction accuracy (NRMSE=0.68) and temporal correlation (r=0.8) between input and decoder output are demonstrated. This approach is more readily applicable to 3D fMRI data as compared to the surface-based model. 

In summary, this work has proposed and developed multivariate and deep learning analysis techniques for brain imaging data in the context of AD with the ultimate goal of improving detection and intervention for early pathological changes in the brain.

Zoom Link: https://umich.zoom.us/meeting/register/tJIpcOigrjIsHtJq_xJ1aboK1T0PdWpTkBP5
*Registration Required

Committee Chair(s):
Dr. Scott J. Peltier and Dr. Douglas C. Noll

]]>
Lecture / Discussion Thu, 01 Sep 2022 11:07:14 -0400 2022-09-26T10:00:00-04:00 2022-09-26T11:00:00-04:00 Off Campus Location Biomedical Engineering Lecture / Discussion BME Ph.D. Defense
Evaluation of Phosphate Treatment and Long-Term RUNX2 Suppression On Adult Human MSC Chondrogenesis and Neo-Cartilage Formation (September 26, 2022 3:15pm) https://events.umich.edu/event/99026 99026-21797474@events.umich.edu Event Begins: Monday, September 26, 2022 3:15pm
Location: Lurie Biomedical Engineering (formerly ATL)
Organized By: Biomedical Engineering

Abstract:
Clinical repair strategies for articular cartilage defects are limited by the inability of the tissue to self-repair, often resulting in post-traumatic osteoarthritis (PTOA). PTOA arises from the degradation of structural cartilage extracellular matrix (ECM) proteins responsible for maintaining articular cartilage mechanics, such as aggrecan and collagen. Current cartilage tissue engineering strategies aim to utilize human-derived cells to regenerate cartilage prior to the onset of PTOA. Limited availability of chondrocytes – the primary cell type in articular cartilage – imposes a need for alternatives. Human mesenchymal stem cells (hMSCs) are a promising solution as they can be found in a variety of tissues and can differentiate into MSC-derived chondrocytes (MdChs). However, MSCs are limited by their inability to produce a stable chondrogenic phenotype and deposit and maintain ECM in long-term culture due to maturation, (hypertrophy) where metalloproteinases cleave collagen II and aggrecan. As a result, MSC-derived cartilage regeneration techniques are not yet suitable for clinical use. The central objective of this thesis is to increase cartilage matrix accumulation for more clinically functional cartilage tissue by increasing matrix deposition and stabilizing the chondrogenic phenotype of MSCs.

We investigated two approaches to increase cartilage ECM accumulation and improve MdCh-based cartilage tissue engineering functional outcomes: inorganic phosphate (Pi) treatment and RUNX2 suppression. First, we found that Pi increased cartilage ECM production, but also increased MdCh hypertrophy, while RUNX2 suppression increased stiffness of neo-cartilage tissues long-term. Finally, we showed that combined treatment of Pi and RUNX2 suppression exhibited reduced MdCh hypertrophy but did not significantly increase matrix accumulation. Overall, this dissertation explores methodologies that promote both cartilage matrix accumulation and reduces cartilage matrix loss during long-term culture to better support the use of MdChs in cartilage defect repair strategies.

Zoom Link: https://umich.zoom.us/j/98189564171 Password: cartilage

Committee Chair: Dr. Rhima Coleman

]]>
Lecture / Discussion Mon, 19 Sep 2022 15:31:39 -0400 2022-09-26T15:15:00-04:00 2022-09-26T16:15:00-04:00 Lurie Biomedical Engineering (formerly ATL) Biomedical Engineering Lecture / Discussion BME PhD Defense
Towards a digital lung for medical research and applications (October 6, 2022 4:30pm) https://events.umich.edu/event/99561 99561-21798343@events.umich.edu Event Begins: Thursday, October 6, 2022 4:30pm
Location: Electrical Engineering and Computer Science Building
Organized By: Biomedical Engineering

Abstract:
Respiratory infections and chronic diseases have been among the top causes of death worldwide for decades, killing over 8 million people annually. Current diagnostic methods for respiratory diseases result in late diagnosis and high rates of underdiagnosis. Further, the wide variability in the patient response to treatment challenges population-based therapies. Personalized medicine arises as a promising alternative to standard practice, but it relies on expensive laboratory and testing infrastructure not available worldwide. This motivates the creation of computational models of the human lungs for in silico research and applications.

In this talk, I will present our group's efforts to construct computational models of the lung for medical research and applications. I will review how computer simulations of alveolar structures informed by micro-computed tomography can change our current understanding of the forces acting on the lung extracellular matrix (ECM). Further, I will discuss a class of microstructurally informed models for predicting the lung tissue response and how they can capture mechanical changes in lung mechanics triggered by ECM remodeling. Finally, I will show our current work on creating personalized virtual lungs and how we can use them to simulate the response of patients under respiratory failure that are connected to mechanical ventilation.

Bio:
Daniel E. Hurtado is an associate professor with the School of Engineering at PUC Chile, and a visiting professor at the Institute for Medical Engineering and Science at MIT. He leads the Computational Medicine Group, an interdisciplinary team that focuses on the creation of personalized computational replicas of the human lungs, with applications in the study of mechanical ventilation and early diagnosis of pulmonary diseases. His work also involves the development of wearable respiratory systems to monitor breathing in athletes and hospital patients. 

Prof. Hurtado received his M.S. and Ph.D. degrees from the California Institute of Technology as a Fulbright fellow. His thesis work made him the recipient of the Robert J. Melosh Medal, presented by Duke University. In 2018, the World Economic Forum selected him as one of the 50 most influential young scientists worldwide under 40 years old for his contributions in research and innovation in biomedical engineering. He is also an elected member for the World Council of Biomechanics.

Zoom:
https://umich.zoom.us/j/91375430500

]]>
Workshop / Seminar Thu, 29 Sep 2022 15:17:19 -0400 2022-10-06T16:30:00-04:00 2022-10-06T17:30:00-04:00 Electrical Engineering and Computer Science Building Biomedical Engineering Workshop / Seminar BME 500 Seminar
Spinal cord stimulation to restore sensation and reduce phantom limb pain after limb amputation (October 20, 2022 4:30pm) https://events.umich.edu/event/98857 98857-21797273@events.umich.edu Event Begins: Thursday, October 20, 2022 4:30pm
Location: Electrical Engineering and Computer Science Building
Organized By: Biomedical Engineering

Abstract:
Despite important advances in the design of prosthetic devices, loss of a limb causes major challenges that often limit participation in activities of daily living. For people with upper-limb amputation, prosthetic adoption rates remain poor and device control is often unintuitive. Those with lower-limb amputation experience impaired balance control, abnormal gait, and an increased rate of falls. Across both groups, upwards of 85% of people also experience debilitating phantom limb pain. All these problems can be attributed, in part, to the loss of sensory feedback from the limb after amputation. In this talk, I will present our research efforts focused on development of devices to stimulate the spinal cord to restore sensory feedback in people with limb amputation. Using devices that are currently implanted in over 50,000 people every year to treat chronic pain, we have demonstrated that spinal cord stimulation can evoke sensations in the missing limb to improve control of prosthetic limbs and reduce phantom limb pain in people with both upper- and lower-limb amputation.

Bio:
Lee Fisher is an Assistant Professor in the Departments of Physical Medicine & Rehabilitation and Bioengineering at the University of Pittsburgh and Director of Education for the Rehab Neural Engineering Labs. Dr. Fisher received his PhD in Biomedical Engineering from Case Western Reserve University, where his research focused on the use of electrical stimulation to restore standing after spinal cord injury. He was a post-doctoral scholar at the University of Pittsburgh before joining the faculty in 2013. Dr. Fisher was the 2021 recipient of the North American Neuromodulation Society’s Kumar New Investigator Award for his research focused on sensory restoration in people with limb amputation. He is a Senior Member of IEEE and Associate Editor of the IEEE Transactions on Neural Systems and Rehabilitation Engineering and Guest Editor of Frontiers in Pain Research.​​​​​​​

Zoom:
https://umich.zoom.us/j/91375430500

]]>
Workshop / Seminar Fri, 16 Sep 2022 08:52:59 -0400 2022-10-20T16:30:00-04:00 2022-10-20T17:30:00-04:00 Electrical Engineering and Computer Science Building Biomedical Engineering Workshop / Seminar BME 500 Seminar
Decoding morphogenic instruction in human microphysiological systems (October 27, 2022 4:30pm) https://events.umich.edu/event/100495 100495-21800009@events.umich.edu Event Begins: Thursday, October 27, 2022 4:30pm
Location: Electrical Engineering and Computer Science Building
Organized By: Biomedical Engineering

Abstract:
Tissue structure and function requires coordination between the physical organization of cells and the genetic programs controlling cell phenotype. Our lab investigates how intersections of chemical and mechanical signals at cell adhesive interfaces function across time and length scales to orchestrate 3D tissue morphogenesis and regulatory signaling. To do so, we develop and apply biomimetic human microphysiological culture systems that incorporate 3D organotypic architectures and permit the study of diverse tissue morphogenic processes associated with human development, regeneration, and pathogenesis with high resolution and biological control. Combining these systems with new molecular tools and microscopy-based methods, we have gained new mechanistic insight into diverse tissue morphogenic events ranging from the genesis and progression of human tuberculosis granulomas to the coordinated assembly and maintenance of human microvascular networks. In this talk, I will highlight these recent advances and detail our recent work establishing a previously unappreciated function for Notch receptors that links adhesive and cytoskeletal processes to transcriptional regulation of cell fate.

Bio:
Matt Kutys is an Assistant Professor in the Department of Cell and Tissue Biology at the University of California San Francisco (UCSF) and is faculty in the Helen Diller Family Comprehensive Cancer Center, the Cardiovascular Research Institute, and the UCSF – UC Berkeley Joint Graduate Program in Bioengineering. Dr. Kutys obtained his BS in Bioengineering from Pennsylvania State University working Dr. William Hancock and he received his PhD in Cell and Developmental Biology from the University of North Carolina Chapel Hill and the National Institutes of Health under Dr. Kenneth Yamada. He was a postdoctoral fellow at Boston University under Dr. Christopher Chen before joining UCSF in 2020. Dr. Kutys is the recipient of an NCI Pathway to Independence Award, a UCSF Program for Breakthrough Biomedical Research Award, and is a Shu Chien Early Career Award Lecturer from the Bioengineering Institute of California. ​​​​​​​

Zoom:
https://umich.zoom.us/j/91375430500

]]>
Workshop / Seminar Thu, 20 Oct 2022 11:22:25 -0400 2022-10-27T16:30:00-04:00 2022-10-27T17:30:00-04:00 Electrical Engineering and Computer Science Building Biomedical Engineering Workshop / Seminar BME 500 Seminar
Magnetic Resonance Imaging for Models of Cardiac Performance (November 3, 2022 4:30pm) https://events.umich.edu/event/100777 100777-21800343@events.umich.edu Event Begins: Thursday, November 3, 2022 4:30pm
Location: Off Campus Location
Organized By: Biomedical Engineering

Abstract:
Medical imaging provides several opportunities to collect data for building patient-specific computational models of the heart. These models estimate performance variables that may not be measured directly (e.g., tissue stress and strain, hemodynamics, or electrical activity). Cardiac magnetic resonance imaging (MRI) can acquire time-resolved images that quantitatively encode structure, function, flow, and remodeling. This talk will summarize recent advances on how these MRI data are acquired and fused using computational models to produce microstructurally anchored measures of patient-specific cardiac performance.

Bio:
Daniel Ennis {he/him} is a Professor in the Department of Radiology at Stanford University. As an MRI scientist for nearly twenty-five years, he has worked to develop advanced translational cardiovascular MRI methods for quantitatively assessing structure, function, flow, and remodeling in both adult and pediatric populations. He began his research career as a Ph.D. student in the Department of Biomedical Engineering at Johns Hopkins University during which time he formed an active collaboration with investigators in the Laboratory of Cardiac Energetics at the National Heart, Lung, and Blood Institute (NIH/NHLBI). Thereafter, he joined the Departments of Radiological Sciences and Cardiothoracic Surgery at Stanford University as a postdoc and began to establish an independent research program with an NIH K99/R00 award focused on “Myocardial Structure, Function, and Remodeling in Mitral Regurgitation.” For ten years he led a group of clinicians and scientists at UCLA working to develop and evaluate advanced cardiovascular MRI exams as PI of several NIH funded studies. In 2018 he returned to the Department of Radiology at Stanford University as faculty in the Radiological Sciences Lab to bolster programs in cardiovascular MRI. He is also the Director of Radiology Research for the Veterans Administration Palo Alto Health Care System where he oversees a growing radiology research program.

Zoom:
https://umich.zoom.us/j/91375430500

]]>
Workshop / Seminar Thu, 27 Oct 2022 11:10:03 -0400 2022-11-03T16:30:00-04:00 2022-11-03T17:30:00-04:00 Off Campus Location Biomedical Engineering Workshop / Seminar BME 500 Seminar
Predicting and quantifying cardiovascular growth, remodeling, and heterogeneity (November 10, 2022 4:30pm) https://events.umich.edu/event/100779 100779-21800344@events.umich.edu Event Begins: Thursday, November 10, 2022 4:30pm
Location: Electrical Engineering and Computer Science Building
Organized By: Biomedical Engineering

Abstract:
Cardiovascular soft tissues serve critical mechanical functions within the body, but pathologic changes to these tissues alter their material properties causing disruption or reduction in function. This loss can be sudden, such as the rupture of an aortic aneurysm, or it can be gradual, such as ventricular hypertrophy and heart failure. In this talk, I will share strategies for predicting and quantifying the temporal and spatial characteristics of cardiovascular soft tissues. First, I will discuss developing and employing a computational model to predict cardiac growth and remodeling. Second, I will expand on experimental testing and analysis techniques for determining the heterogeneous properties of soft tissues. I will close by discussing future applications of these modeling and analyses techniques.

Bio:
Dr. Colleen Witzenburg is an Assistant Professor in the Biomedical Engineering Department at the University of Wisconsin-Madison. She also has an affiliate appointment in the Mechanical Engineering Department, Department of Pediatrics, and is a member of the Cardiovascular Research Center. Dr. Witzenburg earned BS in Mechanical Engineering from Iowa State University and a PhD in Mechanical Engineering from the University of Minnesota. She then completed postdoctoral research at the University of Virginia. Her lab group, the Cardiovascular Biomechanics Lab (cbl.engr.wisc.edu), has been awarded grants from the American Heart Association, the Children’s Heart Foundation, the National Science Foundation, the Wisconsin Alumni Research Foundation, and the UW Institute for Clinical and Translational Research.

Zoom:
https://umich.zoom.us/j/91375430500

]]>
Workshop / Seminar Thu, 27 Oct 2022 11:40:03 -0400 2022-11-10T16:30:00-05:00 2022-11-10T17:30:00-05:00 Electrical Engineering and Computer Science Building Biomedical Engineering Workshop / Seminar BME 500 Seminar
Spatial transcriptomics and cell-free nucleic acids to map host-microbe interactions (November 17, 2022 4:30pm) https://events.umich.edu/event/100783 100783-21800348@events.umich.edu Event Begins: Thursday, November 17, 2022 4:30pm
Location: Electrical Engineering and Computer Science Building
Organized By: Biomedical Engineering

Abstract:
Despite the centrality of microbes to human health, we know very little about how microbes interact with each other and their host. This lack of understanding is in large part due to limitations of tools to measure host-microbe interaction. In the first part of this talk, I will present liquid biopsy technologies to profile host-microbe interaction, with applications in the monitoring of COVID-19 and MIS-C. In the second part of this talk, I will share how we have used spatial transcriptomics to study the pathogenesis of viral myocarditis, and I will present a method to create intricate spatial maps of complex microbial communities.

Bio:
Iwijn De Vlaminck is an associate professor of biomedical engineering at Cornell University. Iwijn’s research is focused on the development of precision medicine technologies to monitor and study infectious and immune related disease. His research has led to noninvasive liquid biopsies to diagnose organ transplant rejection, urinary tract infection, blood-borne infection and complications of stem cell transplantation. He developed methods to spatially map the microbiome and the transcriptome. Iwijn’s research was recognized with the NIH New Innovator Award, the Noyce Foundation Assistant Professorship in the Life Sciences, and a Rainin Foundation Synergy Award. He received teaching excellence awards from the College of Engineering in 2017 and 2022. He is a co-founder of Kanvas Biosciences.

Zoom:
https://umich.zoom.us/j/91375430500

]]>
Workshop / Seminar Thu, 27 Oct 2022 14:07:52 -0400 2022-11-17T16:30:00-05:00 2022-11-17T17:30:00-05:00 Electrical Engineering and Computer Science Building Biomedical Engineering Workshop / Seminar BME 500 Seminar
Engineering Advanced Materials for Neural Regeneration (November 18, 2022 2:30pm) https://events.umich.edu/event/100818 100818-21800387@events.umich.edu Event Begins: Friday, November 18, 2022 2:30pm
Location: Lurie Biomedical Engineering (formerly ATL)
Organized By: Biomedical Engineering

Abstract:
Damage to peripheral nerve and spinal cord tissue can have a devastating impact on the quality of life for individuals suffering from nerve injuries. Our research broadly encompasses analyzing and designing natural-based and electrically conducting biomaterials that can interface with neurons to stimulate and guide nerves to regenerate. This talk will specifically address our work on natural-based biomaterials for both peripheral nerve and spinal cord applications.

To foster peripheral nerve regeneration, we have focused on both “top down” and “bottom up” approaches. For our “top down” approach, we have developed natural acellular tissue grafts created by chemical processing of normal intact nerve tissue to preserve the microarchitecture of the extracellular matrix (ECM) and to eliminate the immune response by removing cell components. This research is the foundation for the Avance Nerve Graft from AxoGen, which is now widely used in clinics for peripheral nerve injuries. In a parallel “bottom up” approach, we have developed advanced hyaluronan-based scaffolds for nerve regeneration applications. Hyaluronic acid (HA; also known as hyaluronan) is a non-sulfated, high molecular weight, glycosaminoglycan found in all mammals; it is a major component of the extracellular matrix in the nervous system and plays a significant role in wound healing and tissue regeneration. Our group has devised novel techniques to process HA into forms for use in peripheral nerve repair applications. For example, we have explored advanced laser-based processes, in situ crystallization, and magnetic particle templating to create microarchitecture within the hyaluronan materials to mimic the native basal lamina of nerve cells and thus to provide physical and chemical guidance features for regenerating axons. These materials have shown promise for supporting peripheral nerve repair after acute transection injury and for promoting regeneration of axons into close proximity of microelectrodes for potential prosthetics applications.

For spinal cord injury (SCI) applications, we have engineered injectable biomaterials for less invasive application in crush injuries, which are the most prominent form of SCI. In this work, we have solubilized decellularized peripheral nerve tissue to create in situ gelling ECM hydrogels. We show that these materials serve as effective therapeutic agents for SCI in rats and are promising delivery agents for cell transplantation applications.

Bio:
Christine E. Schmidt, Ph.D., is the J. Crayton Pruitt Family Professor and Department Chair for the University of Florida Department of Biomedical Engineering. Prior to joining UF in 2013, she was at the University of Texas at Austin in Biomedical Engineering and Chemical Engineering and was one of the founding faculty members of the UT BME Department. 

Dr. Schmidt's research is focused on developing new biomaterials and biomaterial composites (e.g., natural material scaffolds, processed tissues, electronic polymer composites) that can be used to physically guide and stimulate regenerating nerves and the healing of other tissues. Her work is the foundation for the Avance Nerve Repair graft from Axogen and VersaWrap tissue protector from her affiliated start-up company, Alafair Biosciences. Dr. Schmidt has received many major research awards and recognitions, including the Clemson Award for Applied Research from the Society for Biomaterials, induction into the Florida Inventors Hall of Fame, and election to the Florida Academy of Science, Engineering and Medicine of Florida. Dr. Schmidt is a past President for the American Institute for Medical & Biological Engineering (AIMBE). She is a Fellow of AIMBE, the National Academy of Inventors (NAI), the Biomedical Engineering Society (BMES), the American Society for the Advancement of Science (AAAS), the International Academy of Medical and Biological Engineering (IAMBE), and the International Union of Societies for Biomaterials Science and Engineering (FBSE/IUSBSE).

Under Dr. Schmidt’s leadership as Department Chair, UF BME’s undergraduate program first became ABET accredited in Fall 2019 and is now ranked #13 among public BME UG programs (U.S. News & World Report, USNWR). The department’s graduate program is currently ranked #17 among public BME graduate programs by USNWR, climbing more than 20 spots since 2013. Dr. Schmidt has increased the number of women faculty from 2 when she arrived in 2013 to 15 and the number of URM faculty from 1 to 6 (UF BME faculty is now 52% women, 21% URM).

]]>
Lecture / Discussion Wed, 02 Nov 2022 19:59:19 -0400 2022-11-18T14:30:00-05:00 2022-11-18T17:00:00-05:00 Lurie Biomedical Engineering (formerly ATL) Biomedical Engineering Lecture / Discussion Alan J. Hunt Memorial Lecture
Inferring Electromechanical Coupling of the Stomach under Different Gastric States (November 21, 2022 1:00pm) https://events.umich.edu/event/101048 101048-21800727@events.umich.edu Event Begins: Monday, November 21, 2022 1:00pm
Location: Duderstadt Center
Organized By: Biomedical Engineering

Abstract:
A main function of the stomach is to accommodate and break down the ingested food and further push it to the small intestine for nutrient absorption. To carry out this function, gastric smooth muscle cells (SMC) maintain and coordinate their contractions and relaxations across regions of the stomach. The pattern of muscle activity is intrinsically paced by a propagating electrical rhythm initiated by the interstitial cells of Cajal (ICC) and extrinsically regulated by the brain through peripheral nerves innervating the stomach. The ICC-initiated electrical slow wave paces the peristaltic mechanical wave through the active coupling between ICC and SMC (or the electromechanical coupling). The strength of this coupling is up- or down-regulated by the brain through descending vagal nerves, which selectively innervate different types of enteric motor neurons that either excite or inhibit SMC, respectively. The neural control of gastric muscle activity varies across times and conditions to support a wide range of ingestive and digestive processes. In this thesis research, sensors, devices, and signal processing methods were developed for simultaneous recording and real-time analysis of gastric electrical and mechanical activity. Experiments with rats were performed to demonstrate the feasibility of concurrent strain and electrical recordings in both acute and chronic settings. The relationships between the recorded electrical and mechanical activities were evaluated minute-by-minute in terms of their phase, frequency, and amplitude. The electromechanical coupling was stronger and less variable after animals consumed a test meal (or in the fed state) than when the stomach was empty following overnight deprivation of food and drink (or in the fasted state). This finding suggests that the electromechanical coupling may serve as a quantitative biomarker that reports on the real-time neural control of gastric motility, discriminates different gastric states, and by doing so, provides a feedback signal for closed-loop neuromodulation of the stomach. The techniques and findings described in this thesis merit future translational studies to further advance the understanding of gastric physiology and pathophysiology, as well as the diagnosis and treatment of prevailing functional gastrointestinal disorders.

Committee Chair(s):
Dr. Zhongming Liu

Zoom Link: https://umich.zoom.us/j/95286593974
Meeting ID: 952 8659 3974
Passcode: 590590

]]>
Lecture / Discussion Thu, 03 Nov 2022 15:25:04 -0400 2022-11-21T13:00:00-05:00 2022-11-21T14:00:00-05:00 Duderstadt Center Biomedical Engineering Lecture / Discussion Chih Hsuan Tsai
Jonathan Rubin Collegiate Professorship Ceremony (December 5, 2022 1:30pm) https://events.umich.edu/event/101673 101673-21802211@events.umich.edu Event Begins: Monday, December 5, 2022 1:30pm
Location: Palmer Commons
Organized By: Biomedical Engineering

Mary-Ann Mycek, Ph.D.
Professor of Biomedical Engineering
Interim Department Chair, Biomedical Engineering

cordially invites you to a ceremony
celebrating the installation of

Xueding Wang, Ph.D.
as the
Jonathan Rubin Collegiate Professor of Biomedical Engineering

Monday, December 5, 2022
1:30 p.m. - 2:30 p.m. - Ceremony & Lecture

Palmer Commons
Great Lakes South & Central Rooms (4th Floor)
100 Washtenaw Ave, Ann Arbor, MI 48109

Reception to follow from 2:30 p.m. - 3:30 p.m.

]]>
Ceremony / Service Tue, 29 Nov 2022 13:27:27 -0500 2022-12-05T13:30:00-05:00 2022-12-05T14:30:00-05:00 Palmer Commons Biomedical Engineering Ceremony / Service BME Ceremony
Toward an open science ecosystem for neuroimaging (December 8, 2022 4:30pm) https://events.umich.edu/event/101824 101824-21802392@events.umich.edu Event Begins: Thursday, December 8, 2022 4:30pm
Location: Off Campus Location
Organized By: Biomedical Engineering

Abstract:
It is now widely accepted that openness and transparency are keys to improving the reproducibility of scientific research, but many challenges remain to adoption of these practices. I will discuss the growth of an ecosystem for open science within the field of neuroimaging, focusing on platforms for open data sharing and open source tools for reproducible data analysis. I will also discuss the role of the Brain Imaging Data Structure (BIDS), a community standard for data organization, in enabling this open science ecosystem, and will outline the scientific impacts of these resources.
Bio:
Russell A. Poldrack is the Albert Ray Lang Professor in the Department of Psychology, and Director of the Stanford Center for Open and Reproducible Science.  His research uses neuroimaging to understand the brain systems underlying decision making and executive function.  His lab is also engaged in the development of neuroinformatics tools to help improve the reproducibility and transparency of neuroscience, including the Openneuro.org and Neurovault.org data sharing projects and the Cognitive Atlas ontology.​​​​​​​

Zoom:
https://umich.zoom.us/j/91375430500

]]>
Workshop / Seminar Fri, 02 Dec 2022 14:35:11 -0500 2022-12-08T16:30:00-05:00 2022-12-08T17:30:00-05:00 Off Campus Location Biomedical Engineering Workshop / Seminar People in a classroom watching a presentation with the text Biomedical Engineering Seminar Series
Machine Learning-Based Feature Quantification of Clinical High-Frequency Oscillations (December 9, 2022 10:00am) https://events.umich.edu/event/101676 101676-21802214@events.umich.edu Event Begins: Friday, December 9, 2022 10:00am
Location: Lurie Biomedical Engineering (formerly ATL)
Organized By: Biomedical Engineering

Abstract:
The suspense of not knowing when and how a seizure may occur is one of the most exhaustive aspects of an epileptic patient’s life. To address this problem, researchers have been investigating novel biomarkers to gain information about seizure generation and epileptic networks for decades. With modern advancements in recording equipment and computational power, biomarkers that identified from the electrical activity recorded by intracranial electrodes, such as High-Frequency Oscillations (HFOs) have gained traction and are utilized to predict seizure onset zone (SOZ) locations within the brain. However, HFOs are low amplitude waveforms with a time period less than 1 millisecond whose acquisition process can result in noise artifacts. The EEG signals undergo filtering to isolate the HFO events within the 100 – 500 Hz range; a process that can produce false positives due to the occurrence of Gibb’s Phenomenon. Additionally, these filters can also mask the occurrence of artifacts such as head movement and electrical noise. Thus, to address this problem, a machine learning classifier was developed to distinguish events that are clearly artifact from the true HFOs. In this study, logistic regression models were designed to distinguish HFOs naturally occurring in the brain from false positive resulting artifacts amounting to a Positive Predictive Value (PPV) of 84.59%. The correlations of these correctly identified HFOs with inter-patient variability and feature prioritization were also analyzed in a repeated measures study and the significance of each feature was computed. Future directions of this study would follow the estimation of more features from the HFO data to refine the algorithm and improve the precision of identifying true-positive HFO detections. This will heighten the quality of the HFOs being detected in real-time continuous EEG recordings on translation into a clinical environment in the future.

Zoom Link: https://umich.zoom.us/j/96623464149 Meeting ID: 966 2346 4149

Committee Chair(s):
Dr. William Stacey

]]>
Presentation Tue, 29 Nov 2022 14:18:19 -0500 2022-12-09T10:00:00-05:00 2022-12-09T11:00:00-05:00 Lurie Biomedical Engineering (formerly ATL) Biomedical Engineering Presentation BME Master's Defense
Image-guided Transcranial Histotripsy for Brain Tumors (December 14, 2022 11:00am) https://events.umich.edu/event/101679 101679-21802217@events.umich.edu Event Begins: Wednesday, December 14, 2022 11:00am
Location: Lurie Biomedical Engineering (formerly ATL)
Organized By: Biomedical Engineering

Abstract:
The current surgical treatment of malignant brain tumors is invasive and can lead to bleeding and morbid complications. Histotripsy is a noninvasive cavitational ultrasound surgical method that has shown great promise as a noninvasive neurosurgical technology. This dissertation presents image-guided transcranial histotripsy as a potential neurosurgical modality for brain tumors. The first chapter introduces image guidance, brain tumors, the current standard of care, investigative ablation modalities, the mechanism of transcranial histotripsy, and the potential of transcranial histotripsy as a neurosurgical interventional tool. The second chapter discusses the development of a stereotactic transcranial histotripsy targeting system for in vivo murine brain models, The third chapter investigates the magnetic resonance imaging (MRI) analysis and characterization of in vivo features of transcranial histotripsy on murine models. The fourth chapter discusses the first-pass investigation of the blood-brain barrier (BBB) status following transcranial histotripsy in in vivo mice brains. The fifth chapter presents the system error analysis and feasibility of a neuronavigation-guided transcranial histotripsy (NaviTH) system designed for cadaveric models. The final chapter concludes with future work for murine and cadaveric transcranial histotripsy.

Zoom Link: https://umich.zoom.us/j/91504014959
Meeting ID: 915 0401 4959
Password: 0000

Committee Chair(s):
Dr. Zhen Xu

]]>
Lecture / Discussion Tue, 29 Nov 2022 14:31:28 -0500 2022-12-14T11:00:00-05:00 2022-12-14T12:00:00-05:00 Lurie Biomedical Engineering (formerly ATL) Biomedical Engineering Lecture / Discussion BME PhD defense
Computational Hemodynamic Modeling of Pediatric Cardiovascular Diseases (December 16, 2022 10:00am) https://events.umich.edu/event/101921 101921-21802933@events.umich.edu Event Begins: Friday, December 16, 2022 10:00am
Location: Lurie Biomedical Engineering (formerly ATL)
Organized By: Biomedical Engineering

Abstract:
Pediatric cardiovascular disease (CVD) is one of the leading causes of mortality in children. While innovations in pediatric CVD treatment have improved mortality and morbidity, the incidence of residual disease remains high. An increasing level of detail in patients’ diagnostic data has revealed a growing variability in pathology and hemodynamics. Patient-specific hemodynamics are intrinsically linked to the onset and progression of CVD. Therefore, there is a pressing need to improve our understanding of pediatric CVD while considering patient-specific hemodynamics and individualizing treatment plans. 

Computational hemodynamic modeling synergizes patient-specific hemodynamic data with physical and physiological principles to provide a comprehensive description of an individual’s pathology. In this work, computational models are used to study mechanisms contributing to CVD, aid in patient stratification, and aid in surgical planning in three pediatric CVDs: pulmonary arterial hypertension (PAH), renovascular hypertension caused by an abdominal aortic coarctation (AAC), and hypoplastic left heart syndrome (HLHS).

Committee Chair(s):
Dr. David Kohn

Zoom Link: https://umich.zoom.us/j/92042390057
Passcode: modeling

]]>
Presentation Wed, 07 Dec 2022 15:48:17 -0500 2022-12-16T10:00:00-05:00 2022-12-16T11:00:00-05:00 Lurie Biomedical Engineering (formerly ATL) Biomedical Engineering Presentation BME logo on top of a blurred picture of LBME
Resting-State Functional Organization of the Brain in Blindness and Sight Recovery (December 16, 2022 2:00pm) https://events.umich.edu/event/101885 101885-21802611@events.umich.edu Event Begins: Friday, December 16, 2022 2:00pm
Location: North Campus Research Complex Building 10
Organized By: Biomedical Engineering

Abstract:
Reorganization of the human brain after blindness is well-documented, however, subsequent sight restoration can lead to adaptation that is not as well understood. Successful sight restoration therapy must integrate functionally with the visual system for perception to occur. Thus, our study is strongly motivated by the need to understand brain plasticity after regaining vision. In this thesis, I evaluated use of functional magnetic resonance imaging (fMRI) resting-state functional connectivity (rsFC) for vision studies from two angles: 1) from a methodology perspective, I explored the importance of proper data preprocessing on the resulting rsFC outcome, 2) from a neuroscientific perspective, I examined utility of rsFC as a potential metric of blindness and sight restoration.

It has been shown that choice of analysis pipelines can impact the research findings. Therefore, replication studies that aim to reproduce the previously published results are critically necessary. In the first venue of my research, I verified reproducibility of a well-cited published study on ocular blindness using rsFC. By using the original dataset, I utilized another widely used software package to investigate how applying different implementations of the original pipeline or a more rigorous preprocessing stream can alter the outcomes. These alternative workflows changed the distribution of the whole-brain rsFC and functional network densities, reducing the overlap with the original results. Remarkably, the largest rsFC effects appeared to primarily belong to certain connection pairs, irrespective of the pipeline used, likely demonstrating immunity of the larger effects and likely the true results against suboptimal processing. This may highlight the significance of results verification across different computational streams in search of the true findings.

Functional outcome of using Argus II, as the only retinal prosthesis with FDA approval that has been clinically used, can provide an exceptional opportunity to explore brain’s potential for plasticity upon reintroduction of (artificial) vision. Considerable variability in visual performance has been reported across Argus II recipients that remains unexplained. A previous experiment used fMRI to measure tactile-evoked cross-modal responses in visual cortex and reported no significant group-level results between blind and Argus II groups, possibly due to variability in activation baseline across individuals. The rsFC can potentially overcome this issue by providing a more stable metric. Numerous studies have used rsFC to assess cortical reorganization after blindness, nevertheless, it has rarely been utilized to study sight recovery. 

In this study, four resting-state runs from 10 sighted, 10 blind, with severe retinitis pigmentosa, and 7 Argus II subjects were included. The whole-brain ROI-ROI rsFC and some graph theory functional network measures were calculated and compared at the group level. Some quantities decreased after blindness but were not reversed by vision restitution, including visual-visual rsFC, visual-frontal rsFC and some network measures. On the other hand, significant reduction was observed in visual-somatosensory, visual-auditory, visual-motor and visual-association rsFC after blindness that were all returned to the level of sighted individuals in Argus II recipients. These rsFC measures can potentially serve as biomarkers for blindness and sight restoration, in the absence of or as a complement to the behavioral indices.  The proposed metrics can enhance our understanding of variable outcomes among the receivers of sight restorative technologies and enable tracking rehabilitative progress. Future investigation with larger number of test subjects for this rare condition can further unveil the profound ability of our brain to reorganize, following vision restoration.

Committee Chair(s): James Weiland, PhD

Zoom link: https://umich.zoom.us/j/99207257590
Meeting ID: 992 0725 7590
Passcode: 643920

]]>
Presentation Tue, 06 Dec 2022 10:37:24 -0500 2022-12-16T14:00:00-05:00 2022-12-16T15:00:00-05:00 North Campus Research Complex Building 10 Biomedical Engineering Presentation The Biomedical Engineering Logo on a blurred photo of the LBME building at night.
On the in vivo viscoelastic properties of the human lower birth canal during the first stage of labor: secondary analysis of data from the EASE trial (January 11, 2023 10:00am) https://events.umich.edu/event/102845 102845-21805233@events.umich.edu Event Begins: Wednesday, January 11, 2023 10:00am
Location: GG Brown Laboratory
Organized By: Biomedical Engineering

Abstract:
Up to 19% of nulliparous women sustain pelvic muscle injuries while giving vaginal birth. Since these injuries, which are never repaired, can have life-long sequelae including prolapse and incontinence, it seems reasonable to try to prevent them. From a biomechanical perspective, women with unusually stretch-resistant lower birth canals can expect longer labors and a higher risk for muscle and tissue injury. The goals of this dissertation were to (a) quantify the viscoelastic properties of the lower birth canal during the first stage of labor, with special attention to the effects of age and body mass index; (b) identify those at risk for a long second stage of labor, as well as (c) an elevated risk for levator ani injuries. Following an introductory chapter (Chapter 1), we describe the biomechanics of a new device that dilates the lower birth canal (PREP, Materna Medical, LLC., CA) during the first stage of labor, and its use in the ongoing multicenter EASE clinical trial (Chapter 2).

Next, we quantify the normal ranges of lower birth canal resistance to dilation (tension T) in 56 nulliparas during the first stage of labor. We found remarkable variation in birth canal wall tension across all subjects. Specifically, the tension when reaching 55 mm dilation varied 5.5-fold and was 22% greater in older women and correlated strongly with both relaxation during the 5-minute hold and with the maximum tension (r= 0.80, r = 0.59 with p<0.001), thus a reasonable predictor of resistance to lower canal dilation (Chapter 3). The stiffness of the initial 20 s ramp varied 6.8-fold and was 31% higher in older women.

We then demonstrated that the effect of the initial 20 s ramp-and-5 minute hold of PREP insertion on the viscoelastic properties of the lower birth canal can reliably be characterized using a fractional viscoelastic Zener model (FZM). In 61 nullipara, the relative error for the 20 s ramp-and-5 minute hold using the FZM was 8.1 ± 3.6% and provided a relative prediction error in 60 minutes of dilation of 10.0 ± 5.4%. The FZM constant kβ, an analog for the shorter timer constant, was 33% higher in older women (Chapter 4). We then used the fractional Zener viscoelastic constants obtained in Chapter 4 to predict the duration of the second stage of labor and risk for levator ani injury. As labor outcomes are presently unavailable for the PREP subjects due to the ongoing nature of the EASE trial, the model was tested on data from an earlier constant force PILOT dilator study involving 22 healthy women with available labor outcomes. Our prediction model identified both instances of LA injury and predicted 22% of the variation in the second stage of labor duration (Chapter 5). We conclude that the viscoelastic properties of the lower birth canal in the first stage of labor are responsible for nearly a quarter of the variation in second stage duration. When combined with existing obstetric variables, the PREP dilator may prove helpful in the improved management of vaginal delivery.

Zoom Link: https://umich.zoom.us/j/99452208073 Meeting ID: 994 5220 8073 Passcode: 0000

Committee Chair(s): Dr. James Ashton-Miller

]]>
Presentation Thu, 05 Jan 2023 11:07:02 -0500 2023-01-11T10:00:00-05:00 2023-01-11T11:00:00-05:00 GG Brown Laboratory Biomedical Engineering Presentation BME logo over a blurred picture of LBME
Towards defining principles of cell fate plasticity (January 11, 2023 12:00pm) https://events.umich.edu/event/102851 102851-21805240@events.umich.edu Event Begins: Wednesday, January 11, 2023 12:00pm
Location: North Campus Research Complex Building 10
Organized By: Biomedical Engineering

Abstract:
Cell fate plasticity, a cell’s ability to change its identity and function, produces the wide variety of cell types needed to form, maintain, and regenerate all mammalian tissues. All forms of plasticity are tightly regulated to ensure proper tissue function, and when aberrantly activated, can contribute to pathological phenotypes, including impaired tissue repair and tumorigenesis. Understanding how cellular identity is dysregulated during aging, injury, and disease may yield approaches that correct aberrant cell fate transitions with therapeutic applications. This dissertation seeks to characterize the factors that guard cell fate by integrating innovative experimental techniques, including novel cell lines, animal models, and micro-engineered devices, with high-throughput sequencing assays and bioinformatics tools that map molecular information to cellular responses. 

The first three studies provide insights into the mechanisms that preserve the regenerative capacity of muscle stem cells (MuSCs), the resident stem cell population in skeletal muscle. MuSCs lie in a state of mitotic quiescence during homeostasis and activate upon injury to repair damaged tissue while also renewing the quiescent stem cell pool. Over time, MuSCs accumulate intracellular damage that alters metabolic signaling pathways and transcriptional programs supportive of quiescence, blunting the efficient regeneration of skeletal muscle. To further understand how MuSCs acquire dysfunctional molecular programs in old age, we explore the roles of three-dimensional genome organization, an underexplored histone modification (H4K20me1), and a conserved metabolic regulator (Sestrins) in mediating pathological gene expression both at rest and in response to injury. We find that natural aging in MuSCs is accompanied by subtle shifts in global genome architecture and metabolic signaling that are sufficient to induce aberrant regenerative trajectories. 

The fourth and fifth studies describe continuing efforts to develop new models for engineering cell fate. In one study, we identify barriers to cell fate transitions by reprogramming somatic cells such that a specialized cell fate is erased and a new identity is adopted. This process requires spatial remodeling of the epigenetic landscape that is constrained by interactions between peripheral heterochromatin and the protein scaffold that lines the interior of the nucleus. We show that manipulating components of the nuclear scaffold prior to reprogramming alters mechanical properties of the nucleus and peripheral heterochromatin organization, which converge to drive changes in reprogramming dynamics. Finally, we revisit a century-old hypothesis that tumor cells acquire enhanced metastatic potential through fusion with neighboring stromal cells that express pro-metastatic traits such as motility, chemotaxis, and tissue tropism. To investigate this concept, we demonstrate the efficient production of bi-species heterokaryons from human tumor cells and mouse stromal cells in the brain metastatic niche, which allows genetic and metabolic factors to be assigned to their parent cell by species identity. This approach will allow us to identify and inhibit trans-acting factors that promote malignant fate transitions. 

In summary, this thesis provides mechanistic insights into cell fate plasticity in varying health contexts and explores new models for deciphering cell fate regulation.

Committee Chair(s): Dr. Carlos A. Aguilar

Zoom Link: https://umich.zoom.us/meeting/register/tJ0td-2oqDorHNTHn8ikj0vDQx7x7ljfl7uy

]]>
Presentation Thu, 05 Jan 2023 11:44:36 -0500 2023-01-11T12:00:00-05:00 2023-01-11T13:00:00-05:00 North Campus Research Complex Building 10 Biomedical Engineering Presentation BME logo over blurred photo of LBME
Autonomous Medical Robots Guided by Real-Time 3D Imaging (January 12, 2023 3:30pm) https://events.umich.edu/event/102880 102880-21805277@events.umich.edu Event Begins: Thursday, January 12, 2023 3:30pm
Location: Cooley Building
Organized By: Biomedical Engineering

Abstract:
Medical robots can precisely manipulate tools beyond human capabilities and are thus helpful for surgeries involving delicate tissue interactions. When coupled with live 3D imaging, such robots can independently guide surgical instruments in real time. In practice, however, patients receive only limited benefits from such intraprocedural data streams due to the lack of integration between robots and imaging systems with sufficiently high resolution and framerate. In this seminar, I report on work with medical robots that use optical coherence tomography to guide needle insertions for cornea transplantation, enhance surgeon efficiency with live volumetric guidance, and perform autonomous eye imaging. In addition, I discuss using real-time image feedback to adaptively acquire images that break the framerate-resolution barrier during live 3D imaging.

Bio:
Mark Draelos, MD, PhD, is a surgically-trained physician and engineer who develops novel applications of medical robotics and imaging to improve patient care. After finishing Duke University’s Medical Scientist Training Program where he studied biomedical engineering under Prof. Joseph Izatt, Mark completed an internship in general surgery at Duke University Medical Center. Currently, he is an Assistant Professor of Robotics and Ophthalmology at the University of Michigan, where he directs the Image-Guided Medical Robotics Laboratory. Mark has received K99/R00 support from the National Eye Institute.

Zoom:
https://umich.zoom.us/j/91712262512

]]>
Workshop / Seminar Thu, 05 Jan 2023 13:40:07 -0500 2023-01-12T15:30:00-05:00 2023-01-12T16:30:00-05:00 Cooley Building Biomedical Engineering Workshop / Seminar BME classroom set up for a seminar with guests in the audience.
The Mechanics of the Vagina: Deformations, Tears, and Contractions (January 26, 2023 3:30pm) https://events.umich.edu/event/103053 103053-21805781@events.umich.edu Event Begins: Thursday, January 26, 2023 3:30pm
Location: Cooley Building
Organized By: Biomedical Engineering

Abstract:
The vagina is a complex fibromuscular organ with walls that are usually collapsed against each other. The same walls can, however, stretch a great deal during important physiological functions such as conception, pregnancy, and delivery. The remarkable deformations of this organ have not been characterized, despite their impact on women’s health. In this talk, I will offer an overview of the research being conducted in my lab to quantify the unique mechanics and complex microstructure of the vagina by combining advanced experimental, theoretical, and computational methods. The vaginal tissue in the rat model was found to experience very large and highly inhomogeneous deformations in both the active (relaxed) and passive (contracted) states. Over time, the vaginal walls deform very quickly when the loads are first applied and more slowly as the same loads are sustained, revealing their inherent viscoelasticity. Even in the presence of tears, the vagina can undergo large deformations with collagen fibers re-orienting to slow the propagation of tears. Higher contractions occur in the proximal (closer to the cervix) region than in the distal (closer to the introitus) region due to the smooth muscle fiber organization. Data-driven reduced-order modeling techniques are being used to construct in silico models of vaginal deformations, with the accuracy of higher-fidelity models and the speed of simplified models. Future research will explore how mechanical and microstructural properties of the vagina are altered in pathological conditions such as sexual dysfunction, maternal trauma, and pelvic organ prolapse.

Bio:
Raffaella De Vita is a professor and associate department head in the Department of Biomedical Engineering and Mechanics at Virginia Tech. She received her Laurea in Mathematics from La Seconda Università degli Study di Napoli, Italy, in 2000 and her M.S. and Ph.D. from University of Pittsburgh in 2003 and 2005, respectively. She is the recipient of the American Society of Biomechanics President’s award, the NSF CAREER award, the PECASE Award, and several awards for research, teaching, and outreach excellence at Virginia Tech. She is a fellow of the ASME and AIMBE. She has served as an associate editor for the Journal of Elasticity since 2020 and as an associate editor for the Journal of Biomedical Engineering from 2017 to 2022. Her research focuses on determining the relationship between the mechanical behavior and the complex structure of biological systems using theoretical, computational, and experimental methods.

Zoom:
https://umich.zoom.us/j/91712262512

]]>
Workshop / Seminar Mon, 09 Jan 2023 11:21:42 -0500 2023-01-26T15:30:00-05:00 2023-01-26T16:30:00-05:00 Cooley Building Biomedical Engineering Workshop / Seminar BME Seminar
Richard A. Auhll Endowed  Professorship Ceremony & Lecture (January 27, 2023 3:00pm) https://events.umich.edu/event/102468 102468-21804084@events.umich.edu Event Begins: Friday, January 27, 2023 3:00pm
Location: Pierpont Commons
Organized By: Biomedical Engineering

On Friday, January 27 at 3:00 p.m., the College of Engineering will honor Professor Xudong (Sherman) Fan for his appointment to an endowed professorship. Please join Professor Fan, Dean Alec D. Gallimore, Robert J. Vlasic Dean of Engineering, and Biomedical Engineering Professor and Interim Chair, Mary-Ann Myeck, for the lecture and ceremony in the East Conference Room of Pierpont Commons, 2101 Bonisteel Blvd, Ann Arbor, MI.

Professor Fan will be installed as the Richard A. Auhll Professor of Engineering and will present a lecture titled “Breath and body odor analysis for precision healthcare: from benchtop to wearable.” A reception will follow in the East Conference room.

]]>
Ceremony / Service Wed, 21 Dec 2022 10:35:33 -0500 2023-01-27T15:00:00-05:00 2023-01-27T17:00:00-05:00 Pierpont Commons Biomedical Engineering Ceremony / Service Biomedical Engineering logo over a blurred image of LBME.
Biomedical Engineering (BME 500) Seminar Series - Kate Wofford, PhD (February 2, 2023 3:30pm) https://events.umich.edu/event/104034 104034-21808299@events.umich.edu Event Begins: Thursday, February 2, 2023 3:30pm
Location: Cooley Building
Organized By: Biomedical Engineering

Abstract:
Traumatic brain injury (TBI) affects millions of individuals annually resulting in disrupted neuronal circuitry, neurological deficits, and neuroinflammation. Resident and peripheral immune cells interact with damaged neurons after TBI and can contribute to chronic neuroinflammation and neuropathology. Dr. Wofford’s research focuses on understanding and controlling neuro-immune interactions as a therapeutic strategy for TBI. In this talk, Dr. Wofford will describe recent experiments investigating neuro-immune interactions in an advanced translational model of TBI and will describe one method to control inflammation at the cellular level.

Bio:
Dr. Kate Wofford trained with Kara Spiller at Drexel University and Kacy Cullen at the University of Pennsylvania for her Ph.D. in Biomedical Engineering where she characterized acute neuroinflammation after brain injury and developed a strategy to reprogram immune cells. Now, Kate is a Postdoctoral Fellow in the lab of Kacy Cullen at the University of Pennsylvania where she uses advanced preclinical models to study behavioral, neuropathological, and immunological consequences of brain injury. Her work has been recognized with receipt of multiple awards including the K99/R00 Career Development Award, F32 National Research Service Award, Koerner Award, Outstanding Scholar in Neuroscience Award, Interdisciplinary Collaboration and Research Excellence Fellowship, Anthony Marmarou Award, Sanjeev Kumar Memorial Award, and the Wan Shih Translational Research Award.

Zoom:
https://umich.zoom.us/j/91712262512

]]>
Workshop / Seminar Wed, 25 Jan 2023 20:41:13 -0500 2023-02-02T15:30:00-05:00 2023-02-02T16:30:00-05:00 Cooley Building Biomedical Engineering Workshop / Seminar BME Seminar Series
Bioink Development to Advance 3D Bioprinting (February 9, 2023 3:30pm) https://events.umich.edu/event/104390 104390-21808994@events.umich.edu Event Begins: Thursday, February 9, 2023 3:30pm
Location: Cooley Building
Organized By: Biomedical Engineering

Abstract:
Cardiac tissue engineering has emerged to create living, human, cardiac tissue outside the body as a model system in the near term and as a clinical replacement for diseased or damaged cardiac muscle in the long term. My laboratory seeks to understand the intricate interplay between the extracellular matrix and cardiac cell types in vivo to guide cardiac tissue engineering efforts in vitro. In the course of this seminar I will share our most surprising mechanistic insights and describe how they now guide the development of novel bioink formulations that enable 3D bioprinting of complex cardiac tissues.

Bio:
Brenda Ogle is Professor and Head of Biomedical Engineering, Professor of Pediatrics, and Director of the Stem Cell Institute at the University of Minnesota. Her research team investigates the impact of extracellular matrix proteins on stem cell behavior especially in the context of the cardiovascular system. Insights gleaned over the years established mechanistic links between integrin engagement and the activity of critical transcription factors and most recently led to the development of optimized, extracellular matrix-based bioinks for 3D printing of cardiac muscle mimics featured in Newsweek. The primary strength of her laboratory is the ability to span multiple subdisciplines within both basic science (i.e., stem cell biology, cell-cell fusion, and extracellular matrices) and engineering (cytometry, instrumentation, and 3D printing) fields. Her work received funding from the National Institutes of Health, the National Science Foundation, the Department of Defense, the American Heart Association, the Coulter Foundation, Regenerative Medicine Minnesota, and MnDRIVE. She has partnered on research projects with Becton Dickinson, iCyt, 3M and Medtronic. Professor Ogle is an elected fellow of the American Institute for Medical and Biological Engineering and the Biomedical Engineering Society. She has served as a member of the Board of Directors of the Biomedical Engineering Society, as co-chair of the Women’s Faculty Cabinet, UMN and is recipient of the Mullen-Spector-Truax Women’s Leadership Award.

]]>
Workshop / Seminar Wed, 01 Feb 2023 17:06:54 -0500 2023-02-09T15:30:00-05:00 2023-02-09T16:30:00-05:00 Cooley Building Biomedical Engineering Workshop / Seminar BME Seminar Series
Biomechanics of the Femoropopliteal Artery in the Lower Limb (February 16, 2023 3:30pm) https://events.umich.edu/event/104391 104391-21808995@events.umich.edu Event Begins: Thursday, February 16, 2023 3:30pm
Location: Cooley Building
Organized By: Biomedical Engineering

Abstract:
Despite years of technological and pharmacological improvements, failure rates remain high for the lower extremity peripheral arterial disease repairs, particularly when the repair devices cross the knee joint. Though much work has been done investigating the pathological processes associated with this failure, the underlying mechanisms remain insufficiently understood. The main arterial segment within the leg, the femoropopliteal artery, appears to be significantly different from other peripheral arteries due to lower blood flow and large deformations experienced during flexion of the limbs. Understanding the magnitude of these deformations in different postures and arterial segments may help improve repair devices through benchtop and computational studies of device-artery interactions. These studies rely on comprehensive assessments of arterial mechanics and structure and call for innovative ways of accounting for patient demographics and risk factors to deliver realistic results. We will summarize our findings related to the quantification of the biomechanical environment of the lower limb arteries, describe their structure and mechanical properties in the context of age and disease, present in vitro and computational frameworks to evaluate device-artery interactions, and introduce a preclinical animal model to assess the performance of new endovascular and open surgical repairs for the lower extremity.​​​​​​​

Bio:
Dr. Kamenskiy earned his Ph.D. in Engineering Mechanics from the University of Nebraska-Lincoln, and started his academic career as an Assistant Professor in the Department of Surgery at the University of Nebraska Medical Center. After advancing through the academic ranks and receiving tenure in Surgery, Dr. Kamenskiy joined the Department of Biomechanics at the University of Nebraska at Omaha, where he currently serves as Department Chair. In his research, Dr. Kamenskiy integrates Biomechanics and Medicine using in vivo, ex vivo, in vitro, and in silico methods. His lab is interested in vascular mechanophysiology, mechanobiology, and aging and closely collaborates with vascular surgeon-scientists who share the goal of developing practical solutions to improve clinical outcomes for vascular disease patients. Dr. Kamenskiy has assembled one of the largest databases of human artery mechanical, structural, and demographic characteristics that includes well over 1,000 specimens 12 to 99 years old. This database is a unique resource for understanding the complex and diverse pathology of human blood vessels. In addition to studying ex vivo arteries, the team of Dr. Kamenskiy also utilizes cell and organ culture systems and large animal models to explore the disease pathways they observe in human tissues. This research is accompanied by designing, modeling, and testing of new vascular and endovascular repair materials and devices for various vascular pathologies and trauma that his team carries out using bench-top, porcine, human cadaver, and clinical experiments.

Zoom:
https://umich.zoom.us/j/91712262512

]]>
Workshop / Seminar Wed, 01 Feb 2023 17:33:02 -0500 2023-02-16T15:30:00-05:00 2023-02-16T16:30:00-05:00 Cooley Building Biomedical Engineering Workshop / Seminar BME Seminar Series
Dissecting a post-translational modification code in cardiac reprogramming (February 23, 2023 3:30pm) https://events.umich.edu/event/105100 105100-21810752@events.umich.edu Event Begins: Thursday, February 23, 2023 3:30pm
Location: Cooley Building
Organized By: Biomedical Engineering

Abstract:
Cell fate conversion is associated with extensive epigenetic and post translational modifications (PTMs) and architectural changes of sub-organelles and organelles, yet how these events are interconnected remains unknown. We report here the identification of a phosphorylation code in 14-3-3 binding motifs (PC14-3-3) that greatly stimulates induced cardiomyocyte (iCM) formation from fibroblasts. PC14-3-3 was identified in pivotal functional proteins for iCM reprogramming, including transcription factors and epigenetic factors. Akt1 kinase and PP2A phosphatase were a key writer and eraser of the PC14-3-3 code, respectively. PC14-3-3 activation induces iCM formation with the presence of only Tbx5. In contrast, PC14-3-3 inhibition by mutagenesis or inhibitor-mediated code removal abolished reprogramming. We discovered that key PC14-3-3 embedded factors, such as Hdac4, Mef2c, Nrip1, and Foxo1, formed Hdac4 organized inhibitory nuclear condensates. Notably, PC14-3-3 activation disrupted Hdac4 condensates to promote cardiac gene expression. Our study suggests that sub-organelle dynamics regulated by a post-translational modification code could be a general mechanism for stimulating cell reprogramming and organ regeneration.

Bio:
Dr. Zhong Wang is an Associate Professor of Cardiac Surgery, at the University of Michigan Medical School. The long-term goal of the Wang laboratory is to develop heart therapies to effectively prolong and improve the life of patients with cardiovascular disease. The Wang laboratory has made significant progress in four research directions. One research direction is to define the epigenetic mechanism mediated by ATP-dependent chromatin remodeling in cardiac progenitor specification and differentiation. Direction two is to define essential cross-talks between energy metabolism and epigenetics in heart repair and regeneration. Direction three is to identify epigenetic and post-translational modification mechanism and related molecules in stimulating reprogramming of fibroblasts into cardiomyocytes for heart regeneration. And direction four is to explore novel strategies combining optimal cardiovascular cell types and bioengineering/biomaterials for heart cell therapy.

]]>
Workshop / Seminar Fri, 17 Feb 2023 12:33:28 -0500 2023-02-23T15:30:00-05:00 2023-02-23T16:30:00-05:00 Cooley Building Biomedical Engineering Workshop / Seminar BME Seminar Series
Vascular-targeted Nanoparticles to Protect the Endothelium from Immune-mediated Injury (March 9, 2023 3:30pm) https://events.umich.edu/event/105816 105816-21812997@events.umich.edu Event Begins: Thursday, March 9, 2023 3:30pm
Location: Off Campus Location
Organized By: Biomedical Engineering

Abstract: In solid organ transplantation, the host immune system acts to reject the transplanted graft. This process is facilitated at the graft endothelial surface, where inflamed endothelial cells (ECs) upregulate adhesion molecules and recruit effector cells of the host immune system. To combat this dysfunctional inflammation locally and with more impact than globally administered therapies, anti-inflammatory agents can be administered directly to the graft endothelium. We have designed a strategy for local and sustained delivery of these agents using molecularly-targeted polymer nanoparticles (NPs) during a period of ex vivo normothermic machine perfusion (EVNMP) of the organ. I will present several approaches for therapeutic delivery using polymeric NPs as well as strategies to direct NPs using molecular targets to the ECs of interest. We have discovered that rapid accumulation of NPs on ECs relies on both the density and accessibility of the potential ligands, and that these parameters can be measured directly in the relevant human vessel setting. The experiments we have conducted within these platforms are being used to develop a high throughput preclinical approach to optimize immune therapy for local and robust treatment in human organ transplant.

Bio: I joined the faculty at Villanova University in the fall of 2022 in the Department of Chemical and Biological Engineering. I am continuing my research into NP-based therapeutic delivery to human vasculature and integrating these strategies with tissue-engineering to create tools for long-term immune modulation. Specifically, materials that provide support for tissue regrowth while temporarily inhibiting inflammation-related injury, thus reducing the burden of chronic inflammation. My work presented here today was done as a Postdoctoral Fellow in Biomedical Engineering at Yale University as part of Dr. W. Mark Saltzman’s research group. In my graduate work, I developed vascular, tissue engineered constructs using a combination of biological and synthetic materials at the University of Maryland with Dr. John Fisher in Bioengineering.

]]>
Workshop / Seminar Mon, 06 Mar 2023 17:21:36 -0500 2023-03-09T15:30:00-05:00 2023-03-09T16:30:00-05:00 Off Campus Location Biomedical Engineering Workshop / Seminar BME Seminar Series
Automated Design to Engineer Organisms: Scaling up Synthetic Biology to Tackle Humanity's Challenges (March 16, 2023 3:30pm) https://events.umich.edu/event/106090 106090-21813703@events.umich.edu Event Begins: Thursday, March 16, 2023 3:30pm
Location: Cooley Building
Organized By: Biomedical Engineering

Automated Design to Engineer Organisms: Scaling up Synthetic Biology to Tackle Humanity's Challenges

Abstract:
Organism engineering is the bedrock of biotechnology from producing high-value products (enzymes, materials, therapeutics) to developing cell therapies. With the latest techniques in DNA synthesis and assembly, it is now possible to construct large genetic systems with (just about) any DNA sequence of interest, enabling one to engineer sophisticated genetic systems inside cells with many genetic parts. Engineered genetic systems can act as sensors, circuits, and actuators to detect environmental states and autonomously act to change them, for example, probiotic bacteria that sense body temperatures to activate the expression of enzymes that treat metabolic diseases. However, it remains highly challenging to build such genetic systems with high-performance behaviors; there are many “tunable knobs” and inter-dependent interactions that create a “curse of dimensionality” with cryptic (unaccounted for) effects. To overcome these challenges, new approaches are needed that parallel the development of a modern engineering discipline centered around organism engineering.

Specifically, we show that it is now possible to rationally engineer genetic systems by combining predictive models of gene expression together with sequence design algorithms. Our models utilize statistical thermodynamics, kinetics, and machine learning to predict how DNA sequence controls transcription rates, translation rates, mRNA decay rates, gene regulation, and more. Leveraging these model predictions, we automate the design of genetic parts and systems (long DNA sequences) using multi-objective optimization to ensure the engineered organisms have the desired specifications (maximizing target functions, while minimizing undesired behaviors). To develop and test these models, we utilize the latest advances in oligopool synthesis, library-based cloning, and next-generation sequencing to carry out thousands of defined experiments per workflow. We illustrate our rational design approach with several recent applications, including engineering genetic systems to sense-and-respond to human biomarker proteins inside cell-free assays for medical diagnostics and engineering genetic systems to sense-and-respond to TNT inside soil systems for countermine detection.

We have also developed an interactive web-based design platform for engineering organisms, which now has over 10000 registered researchers who have designed over 900,000 genetic systems for diverse biotech applications (medical, industrial, agricultural, defense). The platform provides a “no-code” interface to our suite of predictive models & design algorithms, enabling its broad usage by the community. Altogether, these efforts demonstrate that physiochemical models can indeed predict biological functions with sufficient accuracy to automatically design genetic systems with high performance behaviors.

Bio:
Prof. Howard Salis is an Associate Professor in the Biological Engineering, Chemical Engineering, and Biomedical Engineering departments at Penn State University. He is also a member of the Bioinformatics & Genomics and Molecular, Cellular, & Integrative Biosciences graduate programs. Prof. Salis’ expertise is in the design & engineering of genetic systems in microbial organisms for diverse biotech applications (industrial, medical, agricultural, defense). His lab’s mission is to co-develop a new engineering discipline for biology through the development of predictive models & design algorithms that circumvent the need for trial-and-error experimentation. To develop and test these approaches, his lab carries out thousands of defined experiments per workflow utilizing the latest in oligopool synthesis and next-generation sequencing. Prof. Salis has received the DARPA Young Faculty award and the NSF CAREER award for his achievements. He is also the founder of De Novo DNA, which runs a web-based design platform for engineering organisms, used by over 10000 researchers to design over 900000 genetic systems for diverse biotech applications. Prof. Salis earned his B.S. in Chemical Engineering from Rutgers University (2002) and his Ph.D. in Chemical Engineering from the University of Minnesota (2007). He was a postdoc at UCSF with Chris Voigt (2007-2009). He joined Penn State University in 2010.

Zoom:
https://umich.zoom.us/j/91712262512

]]>
Workshop / Seminar Sat, 11 Mar 2023 21:25:37 -0500 2023-03-16T15:30:00-04:00 2023-03-16T16:30:00-04:00 Cooley Building Biomedical Engineering Workshop / Seminar BME Seminar Series
Dynamic communication networks between regulatory T cells and mesenchymal stromal cells regulate muscle repair and regeneration (March 23, 2023 3:30pm) https://events.umich.edu/event/106476 106476-21814329@events.umich.edu Event Begins: Thursday, March 23, 2023 3:30pm
Location: Cooley Building
Organized By: Biomedical Engineering

Abstract:

Tissue repair and regeneration require a temporally coordinated immune response to clear affected areas and rebuild tissue architecture. To study the dynamic regulation of muscle repair, we generated a time-resolved single-cell RNA sequencing dataset of regulatory T cells (Tregs) and mesenchymal stromal cells (MSCs) in a mouse model of skeletal muscle injury. We built a computational tool to predict the dynamic cellular communication networks between these cell types and found distinct communication pathways during different phases of repair. Using a combination of in vivo CRISPR and genetic mouse models, we validated these interactions and identified novel communication pathways that regulate tissue regeneration.



Short Bio:

Dr. Andrés Muñoz-Rojas is originally from Mexico City. He has a degree in Bioengineering from the University of Pennsylvania. Andrés got his PhD in Biomedical Engineering at Yale University, where he worked with Dr. Kathryn Miller-Jensen using single-cell secretion and transcription technologies to study macrophage polarization in vitro and in tumor microenvironments. He then joined the lab of Diane Mathis at Harvard Medical School as a Postdoctoral Fellow to study tissue immunology and explore the role of Tregs in regulating tissue function.

]]>
Workshop / Seminar Mon, 20 Mar 2023 13:37:03 -0400 2023-03-23T15:30:00-04:00 2023-03-23T16:30:00-04:00 Cooley Building Biomedical Engineering Workshop / Seminar BME Seminar Series
Diagnosing disease on a microchip: Finding nanoscale needles in a nanoscale haystack (March 30, 2023 3:30pm) https://events.umich.edu/event/106711 106711-21814734@events.umich.edu Event Begins: Thursday, March 30, 2023 3:30pm
Location: Cooley Building
Organized By: Biomedical Engineering

Abstract: The transformative growth in microelectronics in the latter half of the 20th century was fueled fundamentally by the ability to miniaturize complex circuits onto chips. The impact of this has been profound– computing is pervasive and portable and communication is instant and global. My research aims to harness this same engineering approach to solve high impact problems in medical diagnostics. To accomplish this goal my lab develops hybrid microchips, where microfluidics are built directly on top of semiconductor chips. In this talk I will focus on recent work at Penn on 'digital asays.' Digital assays — in which ultra-sensitive molecular measurements are made by performing millions of parallel experiments in picoliter droplets — have generated enormous enthusiasm due to their single molecule resolution. These assays have incredible untapped potential for disease diagnostics but are currently confined to laboratory settings due to the instrumentation necessary to generate, control, and measure tens of millions of droplets. To overcome this challenge, we are developing a hybrid microelectronic / microfluidic chip to ‘unlock’ droplet-based assays for mobile use. Our microDroplet Fluorescence Detector (µDFD) takes inspiration from cellular networks, in which phones are identified by their carrier frequency and not their particular location. In collaboration with physicians at The Abramson Cancer Center, we are demonstrating the power of this approach by developing a multiplexed exosome-based diagnostic for the early detection of pancreatic cancer.

Bio: The Issadore lab combines microelectronics, microfluidics, nanomaterials, and machine learning to solve big problems in healthcare. We create miniaturized platforms for the diagnosis of disease, we develop new platforms to manufacture micro and nanomaterials, and we dip our toes into an assortment of other areas where we can leverage our engineering training to improve healthcare. This work requires an interdisciplinary approach in which engineers, scientists, and physicians work together in teams.

]]>
Workshop / Seminar Sat, 25 Mar 2023 18:10:52 -0400 2023-03-30T15:30:00-04:00 2023-03-30T16:30:00-04:00 Cooley Building Biomedical Engineering Workshop / Seminar BME Seminar Series
Hallucinations and objective assessments of deep learning technologies for medical image formation (April 6, 2023 3:30pm) https://events.umich.edu/event/107076 107076-21815261@events.umich.edu Event Begins: Thursday, April 6, 2023 3:30pm
Location: Cooley Building
Organized By: Biomedical Engineering

Abstract:
A variety of deep learning-based image restoration and reconstruction methods, generically referred to as image formation methods, have been proposed for use with biomedical images. It is widely accepted that the assessment and refinement of biomedical imaging technologies should be performed by objective, i.e., task-based, measures of image quality (IQ). However, the objective evaluation of deep learning-based image formation technologies remains largely lacking, despite the breakneck speed at which they are being developed. As such, there is an ever-growing collection of methods whose utility and trustworthiness remains largely unknown. Moreover, such methods have the capability to ‘hallucinate’ false structures, which is of significant concern in medical imaging applications. In this work, we report studies in which the performance of deep learning-based image restoration methods is objectively assessed. The performance of the ideal observer (IO) and common linear numerical observers are quantified, and detection efficiencies are computed to assess the impact of deep learning image formation methods on signal detection performance. The numerical results indicate that, in the cases considered, the application of a deep image formation network can result in a loss of task-relevant information in the image, despite improvement in traditional computer-vision metrics. We also demonstrate that traditional and objective IQ measures can vary in opposite ways as a function of network depth. These results highlight the need for the objective evaluation of IQ for deep image formation technologies and may suggest future avenues for improving the effectiveness of medical imaging applications. 

Bio:
Dr. Mark Anastasio is the Donald Biggar Willett Professor in Engineering and the Head of the Department of Bioengineering at the University of Illinois at Urbana-Champaign (UIUC). Before joining UIUC in 2019, he was a Professor of Biomedical Engineering at Washington University in St. Louis, where he established one of the nation’s first stand-alone PhD programs in imaging science. Dr. Anastasio’s research accomplishments to the fields of biomedical imaging and image science have been numerous and impactful and his general interests broadly address the computational aspects of image formation, modern imaging science, and applied machine learning. He has conducted research in the fields of diffraction tomography, X-ray phase-contrast imaging, and ultrasound tomography. He one of the world’s leading authorities on photoacoustic computed tomography (PACT) and has made numerous and important contributions to development of PACT for over fifteen years. He has published over 175 peer-reviewed journal papers in leading imaging and optical science journals and was the recipient of a National Science Foundation (NSF) CAREER Award to develop image reconstruction methods. He is a Fellow of the American Institute for Medical and Biological Engineering (AIMBE), the International Academy of Medical and Biological Engineering (IAMBE) and the SPIE. He also served as the Chair of the NIH BMIT-B and EITA Study Sections.

Zoom:
https://umich.zoom.us/j/91712262512

]]>
Workshop / Seminar Fri, 31 Mar 2023 14:53:29 -0400 2023-04-06T15:30:00-04:00 2023-04-06T16:30:00-04:00 Cooley Building Biomedical Engineering Workshop / Seminar BME Seminar Series
2023 Biomedical Engineering Symposium with Glenn V. Edmonson Lecture (May 4, 2023 10:00am) https://events.umich.edu/event/107592 107592-21816243@events.umich.edu Event Begins: Thursday, May 4, 2023 10:00am
Location: North Campus Research Complex Building 18
Organized By: Biomedical Engineering

The 2023 Biomedical Engineering Symposium with Glenn V. Edmonson Lecture is intended to build the BME community across campus and honor the legacy of the first graduate chair of the Biomedical Engineering program. These events will provide a forum for BME faculty and students campus-wide along with our collaborators to present current research progress and discuss future research opportunities at the interface of engineering and medicine.

Featuring Glenn V. Edmonson Lecture speaker
Naomi Chesler
Chancellor's Inclusive Excellence Professor
Department of Biomedical Engineering University of California, Irvine
Director of the University of California Irvine
Edwards Lifesciences Foundation
Cardiovascular Innovation & Research Center

The events will take place on Thursday, May 4th, from 10:00 AM - 5:00 PM at NCRC, Bldg 18, Dining Hall. Please RSVP by Thursday, April 27th, 2023.

https://forms.gle/9BivDqH4uh4Wvphn9

Questions: Contact bmesymposium2023@umich.edu

]]>
Lecture / Discussion Fri, 14 Apr 2023 09:58:33 -0400 2023-05-04T10:00:00-04:00 2023-05-04T17:00:00-04:00 North Campus Research Complex Building 18 Biomedical Engineering Lecture / Discussion BME Symposium