Network


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

Hotspot


Dive into the research topics where Joshua C. Doloff is active.

Publication


Featured researches published by Joshua C. Doloff.


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.


Advanced Healthcare Materials | 2013

Core-Shell Hydrogel Microcapsules for Improved Islets Encapsulation

Minglin Ma; Alan Chiu; Gaurav Sahay; Joshua C. Doloff; Nimit Dholakia; Raj Thakrar; Joshua Cohen; Arturo Vegas; Delai Chen; Kaitlin M. Bratlie; Tram T. Dang; Roger L. York; Jennifer Hollister-Lock; Gordon C. Weir; Daniel G. Anderson

Islets microencapsulation holds great promise to treat type 1 diabetes. Currently used alginate microcapsules often have islets protruding outside capsules, leading to inadequate immuno-protection. A novel design of microcapsules with core-shell structures using a two-fluid co-axial electro-jetting is reported. Improved encapsulation and diabetes correction is achieved in a single step by simply confining the islets in the core region of the capsules.


Advanced Materials | 2013

Modular ‘Click-in-Emulsion’ Bone-Targeted Nanogels

Daniel A. Heller; Yair Levi; Jeisa M. Pelet; Joshua C. Doloff; Jasmine Wallas; George W. Pratt; Shan Jiang; Gaurav Sahay; Avi Schroeder; Josh E. Schroeder; Yieu Chyan; Christopher Zurenko; William Querbes; Miguel Manzano; Daniel S. Kohane; Robert Langer; Daniel G. Anderson

A new class of nanogel demonstrates modular biodistribution and affinity for bone. Nanogels, ∼70 nm in diameter and synthesized via an astoichiometric click-chemistry in-emulsion method, controllably display residual, free clickable functional groups. Functionalization with a bisphosphonate ligand results in significant binding to bone on the inner walls of marrow cavities, liver avoidance, and anti-osteoporotic effects.


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.


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.


Science Advances | 2018

Endothelial siRNA delivery in nonhuman primates using ionizable low–molecular weight polymeric nanoparticles

Omar F. Khan; Piotr S. Kowalski; Joshua C. Doloff; Jonathan K. Tsosie; Vasudevan Bakthavatchalu; Caroline Bodi Winn; Jennifer L Haupt; Morgan S Jamiel; Robert Langer; Daniel G. Anderson

Dysfunctional endothelial cells contribute to the pathophysiology of many diseases, including vascular disease, stroke, hypertension, atherosclerosis, organ failure, diabetes, retinopathy, and cancer. Toward the goal of creating a new RNA-based therapy to correct aberrant endothelial cell gene expression in humans, efficient gene silencing in the endothelium of nonhuman primates was achieved by delivering small interfering RNA (siRNA) with 7C1, a low–molecular weight, ionizable polymer that forms nanoparticles. After a single intravenous administration of 1 mg of siRNA per kilogram of animal, 7C1 nanoparticles delivering Tie2 siRNA caused Tie2 mRNA levels to decrease by approximately 80% in the endothelium of the lung. Significant decreases in Tie2 mRNA were also found in the heart, retina, kidney, pancreas, and bone. Blood chemistry and liver function analysis before and after treatment all showed protein and enzyme concentrations within the normal reference ranges. Furthermore, after controlling for siRNA-specific effects, no significant increases in inflammatory cytokine concentrations were found in the serum. Similarly, no gross lesions or significant underlying pathologies were observed after histological examination of nonhuman primate tissues. This study is the first demonstration of endothelial gene silencing in multiple nonhuman primate organs using systemically administered siRNA nanoparticles and highlights the potential of this approach for the treatment of disease in humans.

Collaboration


Dive into the Joshua C. Doloff's collaboration.

Top Co-Authors

Avatar

Daniel G. Anderson

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

Omid Veiseh

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Robert Langer

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Andrew Bader

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
Top Co-Authors

Avatar

Dale L. Greiner

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Michael Chen

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge