Network


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

Hotspot


Dive into the research topics where Nafiseh Masoumi is active.

Publication


Featured researches published by Nafiseh Masoumi.


Biomaterials | 2013

PGS:Gelatin nanofibrous scaffolds with tunable mechanical and structural properties for engineering cardiac tissues.

Mahshid Kharaziha; Mehdi Nikkhah; Su Ryon Shin; Nasim Annabi; Nafiseh Masoumi; Akhilesh K. Gaharwar; Gulden Camci-Unal; Ali Khademhosseini

A significant challenge in cardiac tissue engineering is the development of biomimetic grafts that can potentially promote myocardial repair and regeneration. A number of approaches have used engineered scaffolds to mimic the architecture of the native myocardium tissue and precisely regulate cardiac cell functions. However, previous attempts have not been able to simultaneously recapitulate chemical, mechanical, and structural properties of the myocardial extracellular matrix (ECM). In this study, we utilized an electrospinning approach to fabricate elastomeric biodegradable poly(glycerol sebacate) (PGS):gelatin nanofibrous scaffolds with a wide range of chemical composition, stiffness and anisotropy. Our findings demonstrated that through incorporation of PGS, it is possible to create nanofibrous scaffolds with well-defined anisotropy that mimic the left ventricular myocardium architecture. Furthermore, we studied attachment, proliferation, differentiation and alignment of neonatal rat cardiac fibroblast cells (CFs) as well as protein expression, alignment, and contractile function of cardiomyocyte (CMs) on PGS:gelatin scaffolds with variable amount of PGS. Notably, aligned nanofibrous scaffold, consisting of 33 wt. % PGS, induced optimal synchronous contractions of CMs while significantly enhanced cellular alignment. Overall, our study suggests that the aligned nanofibrous PGS:gelatin scaffold support cardiac cell organization, phenotype and contraction and could potentially be used to develop clinically relevant constructs for cardiac tissue engineering.


Biomaterials | 2014

Tough and flexible CNT-polymeric hybrid scaffolds for engineering cardiac constructs

Mahshid Kharaziha; Su Ryon Shin; Mehdi Nikkhah; Seda Nur Topkaya; Nafiseh Masoumi; Nasim Annabi; Mehmet R. Dokmeci; Ali Khademhosseini

In the past few years, a considerable amount of effort has been devoted toward the development of biomimetic scaffolds for cardiac tissue engineering. However, most of the previous scaffolds have been electrically insulating or lacked the structural and mechanical robustness to engineer cardiac tissue constructs with suitable electrophysiological functions. Here, we developed tough and flexible hybrid scaffolds with enhanced electrical properties composed of carbon nanotubes (CNTs) embedded aligned poly(glycerol sebacate):gelatin (PG) electrospun nanofibers. Incorporation of varying concentrations of CNTs from 0 to 1.5% within the PG nanofibrous scaffolds (CNT-PG scaffolds) notably enhanced fiber alignment and improved the electrical conductivity and toughness of the scaffolds while maintaining the viability, retention, alignment, and contractile activities of cardiomyocytes (CMs) seeded on the scaffolds. The resulting CNT-PG scaffolds resulted in stronger spontaneous and synchronous beating behavior (3.5-fold lower excitation threshold and 2.8-fold higher maximum capture rate) compared to those cultured on PG scaffold. Overall, our findings demonstrated that aligned CNT-PG scaffold exhibited superior mechanical properties with enhanced CM beating properties. It is envisioned that the proposed hybrid scaffolds can be useful for generating cardiac tissue constructs with improved organization and maturation.


Journal of Biomaterials Applications | 2014

Fiber-reinforced hydrogel scaffolds for heart valve tissue engineering

Maryam Eslami; Nihal Engin Vrana; Pinar Zorlutuna; Shilpa Sant; Sungmi Jung; Nafiseh Masoumi; Ramazan Ali Khavari-Nejad; Gholamreza Javadi; Ali Khademhosseini

Heart valve-related disorders are among the major causes of death worldwide. Although prosthetic valves are widely used to treat this pathology, current prosthetic grafts cannot grow with the patient while maintaining normal valve mechanical and hemodynamic properties. Tissue engineering may provide a possible solution to this issue through using biodegradable scaffolds and patients’ own cells. Despite their similarity to heart valve tissue, most hydrogel scaffolds are not mechanically suitable for the dynamic stresses of the heart valve microenvironment. In this study, we integrated electrospun poly(glycerol sebacate) (PGS)–poly(ɛ-caprolactone) (PCL) microfiber scaffolds, which possess enhanced mechanical properties for heart valve engineering, within a hybrid hydrogel made from methacrylated hyaluronic acid and methacrylated gelatin. Sheep mitral valvular interstitial cells were encapsulated in the hydrogel and evaluated in hydrogel-only, PGS–PCL scaffold-only, and composite scaffold conditions. Although the cellular viability and metabolic activity were similar among all scaffold types, the presence of the hydrogel improved the three-dimensional distribution of mitral valvular interstitial cells. As seen by similar values in both the Young’s modulus and the ultimate tensile strength between the PGS–PCL scaffolds and the composites, microfibrous scaffolds preserved their mechanical properties in the presence of the hydrogels. Compared to electrospun or hydrogel scaffolds alone, this combined system may provide a more suitable three-dimensional structure for generating scaffolds for heart valve tissue engineering.


Advanced Healthcare Materials | 2014

Electrospun PGS:PCL microfibers align human valvular interstitial cells and provide tunable scaffold anisotropy.

Nafiseh Masoumi; Benjamin L. Larson; Nasim Annabi; Mahshid Kharaziha; Behnam Zamanian; Kayle Shapero; Alexander T. Cubberley; Gulden Camci-Unal; Keefe B. Manning; John E. Mayer; Ali Khademhosseini

Tissue engineered heart valves (TEHV) can be useful in the repair of congenital or acquired valvular diseases due to their potential for growth and remodeling. The development of biomimetic scaffolds is a major challenge in heart valve tissue engineering. One of the most important structural characteristics of mature heart valve leaflets is their intrinsic anisotropy, which is derived from the microstructure of aligned collagen fibers in the extracellular matrix (ECM). In the present study, a directional electrospinning technique is used to fabricate fibrous poly(glycerol sebacate):poly(caprolactone) (PGS:PCL) scaffolds containing aligned fibers, which resemble native heart valve leaflet ECM networks. In addition, the anisotropic mechanical characteristics of fabricated scaffolds are tuned by changing the ratio of PGS:PCL to mimic the native heart valves mechanical properties. Primary human valvular interstitial cells (VICs) attach and align along the anisotropic axes of all PGS:PCL scaffolds with various mechanical properties. The cells are also biochemically active in producing heart-valve-associated collagen, vimentin, and smooth muscle actin as determined by gene expression. The fibrous PGS:PCL scaffolds seeded with human VICs mimick the structure and mechanical properties of native valve leaflet tissues and would potentially be suitable for the replacement of heart valves in diverse patient populations.


Acta Biomaterialia | 2013

Valvular interstitial cell seeded poly(glycerol sebacate) scaffolds: Toward a biomimetic in vitro model for heart valve tissue engineering

Nafiseh Masoumi; Katherine L. Johnson; M. Christian Howell; George C. Engelmayr

Tissue engineered replacement heart valves may be capable of overcoming the lack of growth potential intrinsic to current non-viable prosthetics, and thus could potentially serve as permanent replacements in the surgical repair of pediatric valvular lesions. However, the evaluation of candidate combinations of cells and scaffolds lacks a biomimetic in vitro model with broadly tunable, anisotropic and elastomeric structural-mechanical properties. Toward establishing such an in vitro model, in the current study, porcine aortic and pulmonary valvular interstitial cells (i.e. biomimetic cells) were cultivated on anisotropic, micromolded poly(glycerol sebacate) scaffolds (i.e. biomimetic scaffolds). Following 14 and 28 days of static culture, cell-seeded scaffolds and unseeded controls were assessed for their mechanical properties, and cell-seeded scaffolds were further characterized by confocal fluorescence and scanning electron microscopy, and by collagen and DNA assays. Poly(glycerol sebacate) micromolding yielded scaffolds with anisotropic stiffnesses resembling those of native valvular tissues in the low stress-strain ranges characteristic of physiologic valvular function. Scaffold anisotropy was largely retained upon cultivation with valvular interstitial cells; while the mechanical properties of unseeded scaffolds progressively diminished, cell-seeded scaffolds either retained or exceeded initial mechanical properties. Retention of mechanical properties in cell-seeded scaffolds paralleled the accretion of collagen, which increased significantly from 14 to 28 days. This study demonstrates that valvular interstitial cells can be cultivated on anisotropic poly(glycerol sebacate) scaffolds to yield biomimetic in vitro models with which clinically relevant cells and future scaffold designs can be evaluated.


Journal of Refractive Surgery | 2012

Finite element analysis of blunt foreign body impact on the cornea after PRK and LASIK

Seyed Jamaleddin Mousavi; Nariman Nassiri; Nafiseh Masoumi; Nader Nassiri; Mercede Majdi-N; Solmaz Farzaneh; Ali R. Djalilian; Gholam Peyman

PURPOSE To investigate the effect of blunt foreign body impact on a human cornea after photorefractive keratectomy (PRK) and LASIK using a simulation model. METHODS Computational simulations were performed using a finite element analysis program (LS-Dyna, Livermore Software Technology Corp). The blunt foreign body was set to impact at the center of the corneal surface models (after PRK and LASIK) with thicknesses of 500, 450, 400, 350, and 300 μm. Corneal rupture was assumed to occur at a peak stress of 9.45 MPa and at a strain of 18%. The foreign body projectile was blunt in shape, made from aluminum, contained plastic-kinematic properties, and had a density of 2700 kg/m(3). RESULTS The projectile was launched at the center of the cornea with velocities ranging from 20 to 60 m/s. The threshold of impact velocities creating rupture in corneal thicknesses of 500, 450, 400, 350, and 300 μm were 33, 32.8, 30.7, 27.9, and 22.8 m/s, respectively, in the PRK model. In the LASIK model, the thresholds creating rupture in the stromal bed of the corneas with thicknesses of 500, 450, 400, 350, and 300 μm were 40, 38.1, 35.6, 31.5, and 26.7 m/s, respectively. The 110-μm corneal flap in the LASIK model ruptured at all velocities. CONCLUSIONS Ruptures occurred at lower velocities in the PRK cornea model than in the corneal stromal bed of the LASIK model following blunt foreign body impact.


Proceedings of the Institution of Mechanical Engineers, Part H: Journal of Engineering in Medicine | 2014

Design and testing of a cyclic stretch and flexure bioreactor for evaluating engineered heart valve tissues based on poly(glycerol sebacate) scaffolds

Nafiseh Masoumi; M. Christian Howell; Katherine L. Johnson; Matthew J Niesslein; Gene Gerber; George C. Engelmayr

Cyclic flexure and stretch are essential to the function of semilunar heart valves and have demonstrated utility in mechanically conditioning tissue-engineered heart valves. In this study, a cyclic stretch and flexure bioreactor was designed and tested in the context of the bioresorbable elastomer poly(glycerol sebacate). Solid poly(glycerol sebacate) membranes were subjected to cyclic stretch, and micromolded poly(glycerol sebacate) scaffolds seeded with porcine aortic valvular interstitial cells were subjected to cyclic stretch and flexure. The results demonstrated significant effects of cyclic stretch on poly(glycerol sebacate) mechanical properties, including significant decreases in effective stiffness versus controls. In valvular interstitial cell-seeded scaffolds, cyclic stretch elicited significant increases in DNA and collagen content that paralleled maintenance of effective stiffness. This work provides a basis for investigating the roles of mechanical loading in the formation of tissue-engineered heart valves based on elastomeric scaffolds.


IEEE Transactions on Biomedical Engineering | 2010

Mathematical Modeling of CSF Pulsatile Hydrodynamics Based on Fluid–Solid Interaction

Nafiseh Masoumi; Dariush Bastani; Siamak Najarian; Fariba Ganji; Farhad Farmanzad; Amir Saeed Seddighi

Intracranial pressure (ICP) is derived from cerebral blood pressure and cerebrospinal fluid (CSF) circulatory dynamics and can be affected in the course of many diseases. Computer analysis of the ICP time pattern plays a crucial role in the diagnosis and treatment of those diseases. This study proposes the application of Linninger et al.s [IEEE Trans. Biomed. Eng. , vol. 52, no. 4, pp. 557-565, Apr. 2005] fluid-solid interaction model of CSF hydrodynamic in ventricular system based on our clinical data from a group of patients with brain parenchyma tumor. The clinical experiments include the arterial blood pressure (ABP), venous blood pressure, and ICP in the subarachnoid space (SAS). These data were used as inputs to the model that predicts the intracranial dynamic phenomena. In addition, the model has been modified by considering CSF pulsatile production rate as the major factor of CSF motion. The approximations of ventricle enlargement, CSF pressure distribution in the ventricular system and CSF velocity magnitude in the aqueduct and foramina were obtained in this study. The observation of reversal flow in the CSF flow pattern due to brain tissue compression is another finding in our investigation. Based on the experimental results, no existence of large transmural pressure differences were found in the brain system. The measured pressure drop in the ventricular system was less than 5 Pa. Moreover, the CSF flow pattern, ICP distribution, and velocity magnitude were in good agreement with the published models and CINE (phase-contrast magnetic resonance imaging) experiments, respectively.


northeast bioengineering conference | 2011

Laser microfabricated poly(glycerol sebacate) scaffolds for heart valve tissue engineering

Nafiseh Masoumi; Katherine L. Johnson; J.T. Zugates; George C. Engelmayr

Toward creating more biomimetic engineered heart valve tissues, in current study we utilized a laser microablation technique to create anisotropic scaffolds comprised of diamond-shaped pores and seeded scaffolds with valvular interstitial cells.


northeast bioengineering conference | 2011

Lipase-resistant poly(glycerol sebacate) via bulk physical entrapment of orlistat

J.T. Zugates; Aurélie Jean; S. Ahmed; Nafiseh Masoumi; George C. Engelmayr

Poly(glycerol sebacate) (PGS) has become one of the most promising new tissue engineering scaffold materials-of-construction. We hypothesized that the lipase inhibitor Orlistat might be suitable for inhibiting lipase-mediated PGS degradation. Here we investigated whether Orlistat can: (1) be physically transported into PGS using 70% (v/v) ethanol in water as a carrier, (2) be stably entrapped within the PGS bulk upon exchange of alcohol for water, and (3) inhibit PGS degradation by lipases. Control 5×5×0.25mm PGS scaffolds comprised of 50 µm struts and identical scaffolds loaded with Orlistat were challenged in vitro by a lipase solution (2000 U/ml). Scaffolds were incubated at 37°C for 1.5, 2, or 3 hours and then assessed by scanning electron microscopy. While evidence of control degradation was apparent after 1.5 h, little degradation was seen in the Orlistat-loaded scaffolds. By 3 hours, while Orlistat-loaded scaffolds began to exhibit modest degradation, controls had undergone rupture of structural elements. Of note, we observed that the path of lipase-mediated degradation was not homogeneous, but appeared to follow pre-existing imperfections in the PGS struts. Results will help guide the design of PGS scaffolds with controllable lipase-resistance.

Collaboration


Dive into the Nafiseh Masoumi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

George C. Engelmayr

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Nasim Annabi

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Benjamin L. Larson

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Katherine L. Johnson

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Aurélie Jean

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Behnam Zamanian

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Gulden Camci-Unal

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

J.T. Zugates

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

John E. Mayer

Boston Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge