Over 8 million people in the US suffer from peripheral arterial disease (PAD), which is characterized by narrowing of the arteries that supply blood flow to the limbs, leading to tissue ischemia. A central feature of PAD is dysfunction of the vascular endothelial cells (ECs) that control vascular reactivity and angiogenesis. We previously demonstrated that ECs derived from human induced pluripotent stem cells (iPSC-ECs) can improve blood perfusion in animals with induced hindlimb ischemia, an experimental model of PAD. However, poor cell survival limited their angiogenic potential. To address this limitation, we seek to develop aligned nanofibrillar collagen scaffolds that mimic the crimped (wavy) structure of native collagen fibrils. In comparison to injectable scaffolds, which lack organized nano-scale structure and mechanical integrity, our preliminary data suggests that aligned nanofibrillar scaffolds provide structural support for guiding the organization of newly formed vessels, directing cell survival, and stimulating angiogenesis. The global hypothesis is that crimped aligned nanofibrillar scaffolds seeded with iPSC-ECs will enhance cell survival, accelerate vascular network formation, and induce angiogenesis in the ischemic limb in small and large animal PAD models. In this resubmission application, Specific Aim 1 will test the hypothesis that crimped aligned nanofibrillar scaffolds, in contrast to randomly oriented scaffolds, will enhance iPSC-EC survival and angiogenesis in vitro. For up to 14 days, cell viability and angiogenesis (migration, vascular network formation, and cytokine production) will be quantitatively compared between iPSC-ECs that are seeded on aligned or randomly oriented scaffolds under hypoxia (1% O2). To validate these findings in vivo, Specific Aim 2 will compare the temporal process of angiogenesis, arteriogenesis, and cell survival after implantation of the iPSC-EC-seeded aligned nanofibrillar scaffold into the ischemic limb. Over 28 days, cell survival, blood perfusion recovery, and formation of new microvasculature will be quantitatively assessed by bioluminescence imaging, laser Doppler blood spectroscopy, and micro computed tomography imaging, respectively, between cells seeded on aligned or randomly oriented scaffolds. Towards clinical translation, Specific Aim 3 will be an exploratory study in which iPSC-ECs seeded on aligned or randomly oriented scaffolds will be implanted into an ovine limb ischemia model. Cell survival will be tracked by. The temporal process of microvessel formation and blood perfusion recovery will be assessed by computed tomography and fluorescence assisted angiography, respectively. Anatomical tunneling of the scaffolds to the ischemic limb will also be examined as a clinically translatable minimally invasive delivery approach. Together, the results of these studies will lead to the development of biomimetic nanofibrillar scaffolds to improve the clinical efficacy of stem cell therapy to PAD patients. We have generated the preliminary scientific components and assembled the expertise to hopefully successfully achieve these goals.

Public Health Relevance

Millions of people in the US suffer from peripheral arterial disease in which blood vessels in the arms or legs become blocked, leading to pain, gangrene, and even limb amputation. Stem cell therapy may be a promising treatment, but poor cell survival limits their ability to help restore blood flow and form new vessels. We aim to study the role of spatially patterned nano-scale biomaterials in enhancing the potential of stem cell-derived endothelial cells to survive and restore blood flow in the setting of peripheral arterial disease. The results of the proposed small and large animal studies will improve the efficacy stem cell therapy for treating peripheral arterial disease.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
1R01HL127113-01A1
Application #
9027928
Study Section
Bioengineering, Technology and Surgical Sciences Study Section (BTSS)
Program Officer
Lee, Albert
Project Start
2016-02-01
Project End
2021-01-31
Budget Start
2016-02-01
Budget End
2017-01-31
Support Year
1
Fiscal Year
2016
Total Cost
Indirect Cost
Name
Stanford University
Department
Surgery
Type
Schools of Medicine
DUNS #
009214214
City
Stanford
State
CA
Country
United States
Zip Code
94304
Nakayama, Karina H; Alcazar, Cynthia; Yang, Guang et al. (2018) Rehabilitative exercise and spatially patterned nanofibrillar scaffolds enhance vascularization and innervation following volumetric muscle loss. NPJ Regen Med 3:16
Zhu, Shoujun; Herraiz, Sonia; Yue, Jingying et al. (2018) 3D NIR-II Molecular Imaging Distinguishes Targeted Organs with High-Performance NIR-II Bioconjugates. Adv Mater 30:e1705799
Foster, Abbygail A; Dewi, Ruby E; Cai, Lei et al. (2018) Protein-engineered hydrogels enhance the survival of induced pluripotent stem cell-derived endothelial cells for treatment of peripheral arterial disease. Biomater Sci 6:614-622
Hou, Luqia; Yang, Guang; Tang, Shibing et al. (2018) Small Molecule Derived From Carboxyethylpyrrole Protein Adducts Promotes Angiogenesis in a Mouse Model of Peripheral Arterial Disease. J Am Heart Assoc 7:e009234
Zamani, Maedeh; Karaca, Esra; Huang, Ngan F (2018) Multicellular Interactions in 3D Engineered Myocardial Tissue. Front Cardiovasc Med 5:147
Zaitseva, Tatiana S; Alcazar, Cynthia; Zamani, Maedeh et al. (2018) Aligned Nanofibrillar Scaffolds for Controlled Delivery of Modified mRNA. Tissue Eng Part A :
Hou, Luqia; Kim, Joseph J; Wanjare, Maureen et al. (2017) Combinatorial Extracellular Matrix Microenvironments for Probing Endothelial Differentiation of Human Pluripotent Stem Cells. Sci Rep 7:6551
Zhong, Yeteng; Ma, Zhuoran; Zhu, Shoujun et al. (2017) Boosting the down-shifting luminescence of rare-earth nanocrystals for biological imaging beyond 1500?nm. Nat Commun 8:737
Wanjare, Maureen; Huang, Ngan F (2017) Regulation of the microenvironment for cardiac tissue engineering. Regen Med 12:187-201
Carcamo-Orive, Ivan; Huang, Ngan F; Quertermous, Thomas et al. (2017) Induced Pluripotent Stem Cell-Derived Endothelial Cells in Insulin Resistance and Metabolic Syndrome. Arterioscler Thromb Vasc Biol 37:2038-2042

Showing the most recent 10 out of 18 publications