Network


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

Hotspot


Dive into the research topics where Andrew Bader is active.

Publication


Featured researches published by Andrew Bader.


Nature Nanotechnology | 2014

In vivo endothelial siRNA delivery using polymeric nanoparticles with low molecular weight

James E. Dahlman; Carmen Barnes; Omar F. Khan; Aude Thiriot; Siddharth Jhunjunwala; Taylor E. Shaw; Yiping Xing; Hendrik B. Sager; Gaurav Sahay; Andrew Bader; Roman L. Bogorad; Hao Yin; Tim Racie; Yizhou Dong; Shan Jiang; Danielle Seedorf; Apeksha Dave; Kamaljeet Singh Sandhu; Matthew J. Webber; Tatiana Novobrantseva; Vera M. Ruda; Abigail K. R. Lytton-Jean; Christopher G. Levins; Brian T. Kalish; Dayna K. Mudge; Mario Perez; Ludmila Abezgauz; Partha Dutta; Lynelle Smith; Klaus Charisse

Dysfunctional endothelium contributes to more diseases than any other tissue in the body. Small interfering RNAs (siRNAs) can help in the study and treatment of endothelial cells in vivo by durably silencing multiple genes simultaneously, but efficient siRNA delivery has so far remained challenging. Here, we show that polymeric nanoparticles made of low-molecular-weight polyamines and lipids can deliver siRNA to endothelial cells with high efficiency, thereby facilitating the simultaneous silencing of multiple endothelial genes in vivo. Unlike lipid or lipid-like nanoparticles, this formulation does not significantly reduce gene expression in hepatocytes or immune cells even at the dosage necessary for endothelial gene silencing. These nanoparticles mediate the most durable non-liver silencing reported so far and facilitate the delivery of siRNAs that modify endothelial function in mouse models of vascular permeability, emphysema, primary tumour growth and metastasis.


Nature Materials | 2015

Size- and shape-dependent foreign body immune response to materials implanted in rodents and non-human primates

Omid Veiseh; Joshua C. Doloff; Minglin Ma; Arturo Vegas; Hok Hei Tam; Andrew Bader; Jie Li; Erin Langan; Jeffrey Wyckoff; Whitney S. Loo; Siddharth Jhunjhunwala; Alan Chiu; Sean Siebert; Katherine Tang; Jennifer Hollister-Lock; Stephanie Aresta-Dasilva; Matthew A. Bochenek; Joshua E. Mendoza-Elias; Yong Wang; Merigeng Qi; Danya M. Lavin; Michael Chen; Nimit Dholakia; Raj Thakrar; Igor Lacík; Gordon C. Weir; Jose Oberholzer; Dale L. Greiner; Robert Langer; Daniel G. Anderson

The efficacy of implanted biomedical devices is often compromised by host recognition and subsequent foreign body responses. Here, we demonstrate the role of the geometry of implanted materials on their biocompatibility in vivo. In rodent and non-human primate animal models, implanted spheres 1.5 mm and above in diameter across a broad spectrum of materials, including hydrogels, ceramics, metals, and plastics, significantly abrogated foreign body reactions and fibrosis when compared to smaller spheres. We also show that for encapsulated rat pancreatic islet cells transplanted into streptozotocin-treated diabetic C57BL/6 mice, islets prepared in 1.5 mm alginate capsules were able to restore blood-glucose control for up to 180 days, a period more than 5-fold longer than for transplanted grafts encapsulated within conventionally sized 0.5-mm alginate capsules. Our findings suggest that the in vivo biocompatibility of biomedical devices can be significantly improved by simply tuning their spherical dimensions.


Nature Medicine | 2016

Long-term glycemic control using polymer-encapsulated human stem cell–derived beta cells in immune-competent mice

Arturo Vegas; Omid Veiseh; Mads Gürtler; Jeffrey R. Millman; Felicia W. Pagliuca; Andrew Bader; Joshua C. Doloff; Jie Li; Michael Chen; Karsten Olejnik; Hok Hei Tam; Siddharth Jhunjhunwala; Erin Langan; Stephanie Aresta-Dasilva; Srujan Gandham; James J. McGarrigle; Matthew A. Bochenek; Jennifer Hollister-Lock; Jose Oberholzer; Dale L. Greiner; Gordon C. Weir; Douglas A. Melton; Robert Langer; Daniel G. Anderson

The transplantation of glucose-responsive, insulin-producing cells offers the potential for restoring glycemic control in individuals with diabetes. Pancreas transplantation and the infusion of cadaveric islets are currently implemented clinically, but these approaches are limited by the adverse effects of immunosuppressive therapy over the lifetime of the recipient and the limited supply of donor tissue. The latter concern may be addressed by recently described glucose-responsive mature beta cells that are derived from human embryonic stem cells (referred to as SC-β cells), which may represent an unlimited source of human cells for pancreas replacement therapy. Strategies to address the immunosuppression concerns include immunoisolation of insulin-producing cells with porous biomaterials that function as an immune barrier. However, clinical implementation has been challenging because of host immune responses to the implant materials. Here we report the first long-term glycemic correction of a diabetic, immunocompetent animal model using human SC-β cells. SC-β cells were encapsulated with alginate derivatives capable of mitigating foreign-body responses in vivo and implanted into the intraperitoneal space of C57BL/6J mice treated with streptozotocin, which is an animal model for chemically induced type 1 diabetes. These implants induced glycemic correction without any immunosuppression until their removal at 174 d after implantation. Human C-peptide concentrations and in vivo glucose responsiveness demonstrated therapeutically relevant glycemic control. Implants retrieved after 174 d contained viable insulin-producing cells.


Nature Biotechnology | 2016

Combinatorial hydrogel library enables identification of materials that mitigate the foreign body response in primates

Arturo Vegas; Omid Veiseh; Joshua C. Doloff; Minglin Ma; Hok Hei Tam; Kaitlin M. Bratlie; Jie Li; Andrew Bader; Erin Langan; Karsten Olejnik; Patrick Fenton; Jeon Woong Kang; Jennifer Hollister-Locke; Matthew A. Bochenek; Alan Chiu; Sean Siebert; Katherine Tang; Siddharth Jhunjhunwala; Stephanie Aresta-Dasilva; Nimit Dholakia; Raj Thakrar; Thema Vietti; Michael Chen; Josh Cohen; Karolina Siniakowicz; Meirigeng Qi; James J. McGarrigle; Stephen Lyle; David M. Harlan; Dale L. Greiner

The foreign body response is an immune-mediated reaction that can lead to the failure of implanted medical devices and discomfort for the recipient. There is a critical need for biomaterials that overcome this key challenge in the development of medical devices. Here we use a combinatorial approach for covalent chemical modification to generate a large library of variants of one of the most widely used hydrogel biomaterials, alginate. We evaluated the materials in vivo and identified three triazole-containing analogs that substantially reduce foreign body reactions in both rodents and, for at least 6 months, in non-human primates. The distribution of the triazole modification creates a unique hydrogel surface that inhibits recognition by macrophages and fibrous deposition. In addition to the utility of the compounds reported here, our approach may enable the discovery of other materials that mitigate the foreign body response.


Embo Molecular Medicine | 2015

Genetic and hypoxic alterations of the microRNA‐210‐ISCU1/2 axis promote iron–sulfur deficiency and pulmonary hypertension

Kevin P. White; Yu Lu; Sofia Annis; Andrew Hale; B. Nelson Chau; James E. Dahlman; Craig Hemann; Alexander R. Opotowsky; Sara O. Vargas; Ivan O. Rosas; Mark A. Perrella; Juan C. Osorio; Kathleen J. Haley; Brian B. Graham; Rahul Kumar; Rajan Saggar; Rajeev Saggar; W. Dean Wallace; David J. Ross; Omar F. Khan; Andrew Bader; Bernadette R. Gochuico; Majed Matar; Kevin Polach; Nicolai M. Johannessen; Haydn M. Prosser; Daniel G. Anderson; Robert Langer; Jay L. Zweier; Laurence A. Bindoff

Iron–sulfur (Fe‐S) clusters are essential for mitochondrial metabolism, but their regulation in pulmonary hypertension (PH) remains enigmatic. We demonstrate that alterations of the miR‐210‐ISCU1/2 axis cause Fe‐S deficiencies in vivo and promote PH. In pulmonary vascular cells and particularly endothelium, hypoxic induction of miR‐210 and repression of the miR‐210 targets ISCU1/2 down‐regulated Fe‐S levels. In mouse and human vascular and endothelial tissue affected by PH, miR‐210 was elevated accompanied by decreased ISCU1/2 and Fe‐S integrity. In mice, miR‐210 repressed ISCU1/2 and promoted PH. Mice deficient in miR‐210, via genetic/pharmacologic means or via an endothelial‐specific manner, displayed increased ISCU1/2 and were resistant to Fe‐S‐dependent pathophenotypes and PH. Similar to hypoxia or miR‐210 overexpression, ISCU1/2 knockdown also promoted PH. Finally, cardiopulmonary exercise testing of a woman with homozygous ISCU mutations revealed exercise‐induced pulmonary vascular dysfunction. Thus, driven by acquired (hypoxia) or genetic causes, the miR‐210‐ISCU1/2 regulatory axis is a pathogenic lynchpin causing Fe‐S deficiency and PH. These findings carry broad translational implications for defining the metabolic origins of PH and potentially other metabolic diseases sharing similar underpinnings.


Nature Materials | 2017

Colony stimulating factor-1 receptor is a central component of the foreign body response to biomaterial implants in rodents and non-human primates

Joshua C. Doloff; Omid Veiseh; Arturo Vegas; Hok Hei Tam; Shady Farah; Minglin Ma; Jie Li; Andrew Bader; Alan Chiu; Atieh Sadraei; Stephanie Aresta-Dasilva; Marissa Griffin; Siddharth Jhunjhunwala; Matthew J. Webber; Sean Siebert; Katherine Tang; Michael Chen; Erin Langan; Nimit Dholokia; Raj Thakrar; Meirigeng Qi; Jose Oberholzer; Dale L. Greiner; Robert Langer; Daniel G. Anderson

Host recognition and immune-mediated foreign body response (FBR) to biomaterials can compromise the performance of implanted medical devices. To identify key cell and cytokine targets, here we perform in-depth systems analysis of innate and adaptive immune system responses to implanted biomaterials in rodents and non-human primates. While macrophages are indispensable to the fibrotic cascade, surprisingly neutrophils and complement are not. Macrophages, via CXCL13, lead to downstream B cell recruitment, which further potentiated fibrosis, as confirmed by B cell knock out and CXCL13 neutralization. Interestingly, Colony Stimulating Factor-1 Receptor (CSF1R) is significantly increased following implantation of multiple biomaterial classes: ceramic, polymer, and hydrogel. Its inhibition, like macrophage depletion, leads to complete loss of fibrosis, but spares other macrophage functions such as wound healing, ROS production, and phagocytosis. Our results indicate targeting CSF1R may allow for a more selective method of fibrosis inhibition, and improve biomaterial biocompatibility without the need for broad immunosuppression.


PLOS ONE | 2015

Neutrophil Responses to Sterile Implant Materials

Siddharth Jhunjhunwala; Stephanie Aresta-Dasilva; Katherine Tang; David Alvarez; Matthew J. Webber; Benjamin C. Tang; Danya M. Lavin; Omid Veiseh; Joshua C. Doloff; Suman Bose; Arturo Vegas; Minglin Ma; Gaurav Sahay; Alan Chiu; Andrew Bader; Erin Langan; Sean Siebert; Jie Li; Dale L. Greiner; Peter E. Newburger; Ulrich H. von Andrian; Robert Langer; Daniel G. Anderson

In vivo implantation of sterile materials and devices results in a foreign body immune response leading to fibrosis of implanted material. Neutrophils, one of the first immune cells to be recruited to implantation sites, have been suggested to contribute to the establishment of the inflammatory microenvironment that initiates the fibrotic response. However, the precise numbers and roles of neutrophils in response to implanted devices remains unclear. Using a mouse model of peritoneal microcapsule implantation, we show 30–500 fold increased neutrophil presence in the peritoneal exudates in response to implants. We demonstrate that these neutrophils secrete increased amounts of a variety of inflammatory cytokines and chemokines. Further, we observe that they participate in the foreign body response through the formation of neutrophil extracellular traps (NETs) on implant surfaces. Our results provide new insight into neutrophil function during a foreign body response to peritoneal implants which has implications for the development of biologically compatible medical devices.


Nano Letters | 2017

Microfluidic Fabrication of Colloidal Nanomaterials-Encapsulated Microcapsules for Biomolecular Sensing

Xi Xie; Weixia Zhang; Alireza Abbaspourrad; Jiyoung Ahn; Andrew Bader; Suman Bose; Arturo Vegas; Jiaqi Lin; Jun Tao; Tian Hang; Hyomin Lee; Nicole M. Iverson; Gili Bisker; Linxian Li; Michael S. Strano; David A. Weitz; Daniel G. Anderson

Implantable sensors that detect biomarkers in vivo are critical for early disease diagnostics. Although many colloidal nanomaterials have been developed into optical sensors to detect biomolecules in vitro, their application in vivo as implantable sensors is hindered by potential migration or clearance from the implantation site. One potential solution is incorporating colloidal nanosensors in hydrogel scaffold prior to implantation. However, direct contact between the nanosensors and hydrogel matrix has the potential to disrupt sensor performance. Here, we develop a hollow-microcapsule-based sensing platform that protects colloidal nanosensors from direct contact with hydrogel matrix. Using microfluidics, colloidal nanosensors were encapsulated in polyethylene glycol microcapsules with liquid cores. The microcapsules selectively trap the nanosensors within the core while allowing free diffusion of smaller molecules such as glucose and heparin. Glucose-responsive quantum dots or gold nanorods or heparin-responsive gold nanorods were each encapsulated. Microcapsules loaded with these sensors showed responsive optical signals in the presence of target biomolecules (glucose or heparin). Furthermore, these microcapsules can be immobilized into biocompatible hydrogel as implantable devices for biomolecular sensing. This technique offers new opportunities to extend the utility of colloidal nanosensors from solution-based detection to implantable device-based detection.


Advanced Healthcare Materials | 2017

A Facile and Versatile Method to Endow Biomaterial Devices with Zwitterionic Surface Coatings

Volkan Yesilyurt; Omid Veiseh; Joshua C. Doloff; Jie Li; Suman Bose; Xi Xie; Andrew Bader; Michael Chen; Matthew J. Webber; Arturo Vegas; Robert Langer; Daniel G. Anderson

The surface modification of implantable biomaterials with zwitterionic phosphorylcholine polymer is demonstrated through mussel-mimetic catecholamine polymer thin films. Using this method, the surfaces of alginate hydrogel microspheres and polystyrene microbeads, a model material known to produce robust foreign body responses and fibrosis, are successfully modified to reduce the tissue reaction by reducing the fibrosis in immunocompetent C57BL/6J mice.


Nature Biomedical Engineering | 2018

Alginate encapsulation as long-term immune protection of allogeneic pancreatic islet cells transplanted into the omental bursa of macaques

Matthew A. Bochenek; Omid Veiseh; Arturo Vegas; James J. McGarrigle; Meirigeng Qi; Enza Marchese; Mustafa Omami; Joshua C. Doloff; Joshua E. Mendoza-Elias; Mohammad Nourmohammadzadeh; A. Khan; Chun Chieh Yeh; Yuan Xing; Douglas Isa; Sofia Ghani; Jie Li; Casey Landry; Andrew Bader; Karsten Olejnik; Michael Chen; Jennifer Hollister-Lock; Yong Wang; Dale L. Greiner; Gordon C. Weir; Berit L. Strand; Anne Mari Rokstad; Igor Lacík; Robert Langer; Daniel G. Anderson; Jose Oberholzer

The transplantation of pancreatic islet cells could restore glycaemic control in patients with type 1 diabetes. Microspheres for islet encapsulation have enabled long-term glycaemic control in rodent models of diabetes; however, humans transplanted with equivalent microsphere formulations have experienced only transient islet graft function owing to a vigorous foreign-body response (FBR), to pericapsular fibrotic overgrowth (PFO) and, in upright bipedal species, to the sedimentation of the microspheres within the peritoneal cavity. Here, we report the results of the testing in non-human primate (NHP) models of seven alginate formulations that were efficacious in rodents, including three that led to transient islet graft function in clinical trials. All formulations elicited significant FBR and PFO 1 month post implantation; however, three chemically modified, immune-modulating alginate formulations elicited a reduced FBR. In conjunction with a minimally invasive transplantation technique into the bursa omentalis of NHPs, the most promising chemically modified alginate derivative (Z1-Y15) protected viable and glucose-responsive allogeneic islets for 4 months without the need for immunosuppression. Chemically modified alginate formulations may enable the long-term transplantation of islets for the correction of insulin deficiency.Transplantation of pancreatic islet cells encapsulated in alginate microspheres into the omental bursa of the peritoneal cavity of NHPs significantly reduces FBRs and extends the longevity of the cells.

Collaboration


Dive into the Andrew Bader's collaboration.

Top Co-Authors

Avatar

Daniel G. Anderson

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Robert Langer

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Arturo Vegas

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

James E. Dahlman

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Jie Li

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Joshua C. Doloff

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Omar F. Khan

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Omid Veiseh

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew Hale

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge