Network


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

Hotspot


Dive into the research topics where Panagiotis Maghsoudlou is active.

Publication


Featured researches published by Panagiotis Maghsoudlou.


Biomaterials | 2012

A rat decellularized small bowel scaffold that preserves villus-crypt architecture for intestinal regeneration

Giorgia Totonelli; Panagiotis Maghsoudlou; Massimo Garriboli; Johannes Riegler; Giuseppe Orlando; Alan J. Burns; Nj Sebire; Virpi V. Smith; Jonathan M. Fishman; Marco Ghionzoli; Mark Turmaine; Martin A. Birchall; Anthony Atala; Shay Soker; Mark F. Lythgoe; Alexander M. Seifalian; Agostino Pierro; Simon Eaton; Paolo De Coppi

Management of intestinal failure remains a clinical challenge and total parenteral nutrition, intestinal elongation and/or transplantation are partial solutions. In this study, using a detergent-enzymatic treatment (DET), we optimize in rats a new protocol that creates a natural intestinal scaffold, as a base for developing functional intestinal tissue. After 1 cycle of DET, histological examination and SEM and TEM analyses showed removal of cellular elements with preservation of the native architecture and connective tissue components. Maintenance of biomechanical, adhesion and angiogenic properties were also demonstrated strengthen the idea that matrices obtained using DET may represent a valid support for intestinal regeneration.


Biomaterials | 2013

Discarded human kidneys as a source of ECM scaffold for kidney regeneration technologies

Giuseppe Orlando; Christopher Booth; Zhan Wang; Giorgia Totonelli; Christina L. Ross; Emma Moran; Marcus Salvatori; Panagiotis Maghsoudlou; Mark Turmaine; Ginger T. DeLario; Yousef Al-Shraideh; Umar Farooq; Alan C. Farney; Jeffrey Rogers; Samy S. Iskandar; Alan J. Burns; Frank C. Marini; Paolo De Coppi; Robert J. Stratta; Shay Soker

In the United States, more than 2600 kidneys are discarded annually, from the total number of kidneys procured for transplant. We hypothesized that this organ pool may be used as a platform for renal bioengineering and regeneration research. We previously showed that decellularization of porcine kidneys yields renal extracellular matrix (ECM) scaffolds that maintain their basic components, support cell growth and welfare in vitro and in vivo, and show an intact vasculature that, when such scaffolds are implanted in vivo, is able to sustain physiological blood pressure. The purpose of the current study was to test if the same strategy can be applied to discarded human kidneys in order to obtain human renal ECM scaffolds. The results show that the sodium dodecylsulfate-based decellularization protocol completely cleared the cellular compartment in these kidneys, while the innate ECM framework retained its architecture and biochemical properties. Samples of human renal ECM scaffolds stimulated angiogenesis in a chick chorioallantoic membrane assay. Importantly, the innate vascular network in the human renal ECM scaffolds retained its compliance. Collectively, these results indicate that discarded human kidneys are a suitable source of renal scaffolds and their use for tissue engineering applications may be more clinically applicable than kidneys derived from animals.


Scientific Reports | 2015

Decellularized human liver as a natural 3D-scaffold for liver bioengineering and transplantation.

Giuseppe Mazza; Krista Rombouts; Andrew R. Hall; Luca Urbani; Tu Vinh Luong; W. Al-Akkad; L. Longato; David A. Brown; Panagiotis Maghsoudlou; Amar P. Dhillon; Barry J. Fuller; Brian Davidson; Kevin Moore; Dipok Kumar Dhar; Paolo De Coppi; Massimo Malago; Massimo Pinzani

Liver synthetic and metabolic function can only be optimised by the growth of cells within a supportive liver matrix. This can be achieved by the utilisation of decellularised human liver tissue. Here we demonstrate complete decellularization of whole human liver and lobes to form an extracellular matrix scaffold with a preserved architecture. Decellularized human liver cubic scaffolds were repopulated for up to 21 days using human cell lines hepatic stellate cells (LX2), hepatocellular carcinoma (Sk-Hep-1) and hepatoblastoma (HepG2), with excellent viability, motility and proliferation and remodelling of the extracellular matrix. Biocompatibility was demonstrated by either omental or subcutaneous xenotransplantation of liver scaffold cubes (5 × 5 × 5 mm) into immune competent mice resulting in absent foreign body responses. We demonstrate decellularization of human liver and repopulation with derived human liver cells. This is a key advance in bioartificial liver development.


Stem Cells and Development | 2011

Amniotic fluid stem cells are cardioprotective following acute myocardial infarction.

Sveva Bollini; King K. Cheung; Johannes Riegler; Xuebin Dong; Nicola Smart; Marco Ghionzoli; S Loukogeorgakis; Panagiotis Maghsoudlou; Karina N. Dubé; Paul R. Riley; Mark F. Lythgoe; P De Coppi

In recent years, various types of stem cells have been characterized and their potential for cardiac regeneration has been investigated. We have previously described the isolation of broadly multipotent cells from amniotic fluid, defined as amniotic fluid stem (AFS) cells. The aim of this study was to investigate the therapeutic potential of human AFS cells (hAFS) in a model of acute myocardial infarction. Wistar rats underwent 30 min of ischemia by ligation of the left anterior descending coronary artery, followed by administration of hAFS cells and 2 h of reperfusion. Infarct size was assessed by 2,3,5-triphenyltetrazolium chloride staining and planimetry. hAFS cells were also analyzed by enzyme-linked immunosorbent assay to detect secretion of putative paracrine factors, such as the actin monomer-binding protein thymosin β4 (Tβ4). The systemic injection of hAFS cells and their conditioned medium (hAFS-CM) was cardioprotective, improving myocardial cell survival and decreasing the infarct size from 53.9%±2.3% (control animals receiving phosphate-buffered saline injection) to 40.0%±3.0% (hAFS cells) and 39.7%±2.5% (hAFS-CM, P<0.01). In addition, hAFS cells were demonstrated to secrete Tβ4, previously shown to be both cardioprotective and proangiogenic. Our results suggest that AFS cells have therapeutic potential in the setting of acute myocardial infarction, which may be mediated through paracrine effectors such as Tβ4. Therefore, AFS cells might represent a novel source for cell therapy and cell transplantation strategies in repair following ischemic heart disease, with a possible paracrine mechanism of action and a potential molecular candidate for acute cardioprotection.


World Journal of Gastroenterology | 2012

Esophageal tissue engineering: A new approach for esophageal replacement

Giorgia Totonelli; Panagiotis Maghsoudlou; Jonathan M. Fishman; Giuseppe Orlando; Tahera Ansari; Paul Sibbons; Martin A. Birchall; Agostino Pierro; Simon Eaton; Paolo De Coppi

A number of congenital and acquired disorders require esophageal tissue replacement. Various surgical techniques, such as gastric and colonic interposition, are standards of treatment, but frequently complicated by stenosis and other problems. Regenerative medicine approaches facilitate the use of biological constructs to replace or regenerate normal tissue function. We review the literature of esophageal tissue engineering, discuss its implications, compare the methodologies that have been employed and suggest possible directions for the future. Medline, Embase, the Cochrane Library, National Research Register and ClinicalTrials.gov databases were searched with the following search terms: stem cell and esophagus, esophageal replacement, esophageal tissue engineering, esophageal substitution. Reference lists of papers identified were also examined and experts in this field contacted for further information. All full-text articles in English of all potentially relevant abstracts were reviewed. Tissue engineering has involved acellular scaffolds that were either transplanted with the aim of being repopulated by host cells or seeded prior to transplantation. When acellular scaffolds were used to replace patch and short tubular defects they allowed epithelial and partial muscular migration whereas when employed for long tubular defects the results were poor leading to an increased rate of stenosis and mortality. Stenting has been shown as an effective means to reduce stenotic changes and promote cell migration, whilst omental wrapping to induce vascularization of the construct has an uncertain benefit. Decellularized matrices have been recently suggested as the optimal choice for scaffolds, but smart polymers that will incorporate signalling to promote cell-scaffold interaction may provide a more reproducible and available solution. Results in animal models that have used seeded scaffolds strongly suggest that seeding of both muscle and epithelial cells on scaffolds prior to implantation is a prerequisite for complete esophageal replacement. Novel approaches need to be designed to allow for peristalsis and vascularization in the engineered esophagus. Although esophageal tissue engineering potentially offers a real alternative to conventional treatments for severe esophageal disease, important barriers remain that need to be addressed.


Biomaterials | 2013

Preservation of micro-architecture and angiogenic potential in a pulmonary acellular matrix obtained using intermittent intra-tracheal flow of detergent enzymatic treatment

Panagiotis Maghsoudlou; Fanourios Georgiades; Athanasios Tyraskis; Giorgia Totonelli; S Loukogeorgakis; Giuseppe Orlando; Panicos Shangaris; Peggy Lange; Jean-Marie Delalande; Alan J. Burns; Angelo Cenedese; Nj Sebire; Mark Turmaine; Brogan Guest; John F. Alcorn; Anthony Atala; Martin A. Birchall; Martin J. Elliott; Simon Eaton; Agostino Pierro; Thomas W. Gilbert; Paolo De Coppi

Tissue engineering of autologous lung tissue aims to become a therapeutic alternative to transplantation. Efforts published so far in creating scaffolds have used harsh decellularization techniques that damage the extracellular matrix (ECM), deplete its components and take up to 5 weeks to perform. The aim of this study was to create a lung natural acellular scaffold using a method that will reduce the time of production and better preserve scaffold architecture and ECM components. Decellularization of rat lungs via the intratracheal route removed most of the nuclear material when compared to the other entry points. An intermittent inflation approach that mimics lung respiration yielded an acellular scaffold in a shorter time with an improved preservation of pulmonary micro-architecture. Electron microscopy demonstrated the maintenance of an intact alveolar network, with no evidence of collapse or tearing. Pulsatile dye injection via the vasculature indicated an intact capillary network in the scaffold. Morphometry analysis demonstrated a significant increase in alveolar fractional volume, with alveolar size analysis confirming that alveolar dimensions were maintained. Biomechanical testing of the scaffolds indicated an increase in resistance and elastance when compared to fresh lungs. Staining and quantification for ECM components showed a presence of collagen, elastin, GAG and laminin. The intratracheal intermittent decellularization methodology could be translated to sheep lungs, demonstrating a preservation of ECM components, alveolar and vascular architecture. Decellularization treatment and methodology preserves lung architecture and ECM whilst reducing the production time to 3 h. Cell seeding and in vivo experiments are necessary to proceed towards clinical translation.


Annals of Surgery | 2016

The human pancreas as a source of protolerogenic extracellular matrix scaffold for a new-generation bioartificial endocrine pancreas

Andrea Peloso; Luca Urbani; Paolo Cravedi; Ravi Katari; Panagiotis Maghsoudlou; Mario Enrique Alvarez Fallas; Valeria Sordi; Antonio Citro; Carolina Purroy; John P. McQuilling; Sivanandane Sittadjody; Alan C. Farney; Samy S. Iskandar; Joao Paulo Zambon; Jeffrey Rogers; Robert J. Stratta; Emmanuel C. Opara; Lorenzo Piemonti; Cristina M. Furdui; Shay Soker; Paolo De Coppi; Giuseppe Orlando

Objectives: Our study aims at producing acellular extracellular matrix scaffolds from the human pancreas (hpaECMs) as a first critical step toward the production of a new-generation, fully human-derived bioartificial endocrine pancreas. In this bioartificial endocrine pancreas, the hardware will be represented by hpaECMs, whereas the software will consist in the cellular compartment generated from patients own cells. Background: Extracellular matrix (ECM)-based scaffolds obtained through the decellularization of native organs have become the favored platform in the field of complex organ bioengineering. However, the paradigm is now switching from the porcine to the human model. Methods: To achieve our goal, human pancreata were decellularized with Triton-based solution and thoroughly characterized. Primary endpoints were complete cell and DNA clearance, preservation of ECM components, growth factors and stiffness, ability to induce angiogenesis, conservation of the framework of the innate vasculature, and immunogenicity. Secondary endpoint was hpaECMs’ ability to sustain growth and function of human islet and human primary pancreatic endothelial cells. Results: Results show that hpaECMs can be successfully and consistently produced from human pancreata and maintain their innate molecular and spatial framework and stiffness, and vital growth factors. Importantly, hpaECMs inhibit human naïve CD4+ T-cell expansion in response to polyclonal stimuli by inducing their apoptosis and promoting their conversion into regulatory T cells. hpaECMs are cytocompatible and supportive of representative pancreatic cell types. Discussion: We, therefore, conclude that hpaECMs has the potential to become an ideal platform for investigations aiming at the manufacturing of a regenerative medicine-inspired bioartificial endocrine pancreas.


Journal of Visualized Experiments | 2013

A decellularization methodology for the production of a natural acellular intestinal matrix.

Panagiotis Maghsoudlou; Giorgia Totonelli; Stavros Loukogeorgakis; Simon Eaton; Paolo De Coppi

Successful tissue engineering involves the combination of scaffolds with appropriate cells in vitro or in vivo. Scaffolds may be synthetic, naturally-derived or derived from tissues/organs. The latter are obtained using a technique called decellularization. Decellularization may involve a combination of physical, chemical, and enzymatic methods. The goal of this technique is to remove all cellular traces whilst maintaining the macro- and micro-architecture of the original tissue. Intestinal tissue engineering has thus far used relatively simple scaffolds that do not replicate the complex architecture of the native organ. The focus of this paper is to describe an efficient decellularization technique for rat small intestine. The isolation of the small intestine so as to ensure the maintenance of a vascular connection is described. The combination of chemical and enzymatic solutions to remove the cells whilst preserving the villus-crypt axis in the luminal aspect of the scaffold is also set out. Finally, assessment of produced scaffolds for appropriate characteristics is discussed.


Seminars in Pediatric Surgery | 2014

Tissue engineering of the esophagus

Panagiotis Maghsoudlou; Simon Eaton; Paolo De Coppi

Esophageal atresia occurs in 1 out of 3000 births. Current treatments involve esophageal replacement by using more distal parts of the gastrointestinal tract, such as the stomach, jejunum, and colon. Significant complications are associated with each treatment option. Tissue engineering may provide a therapeutic alternative for esophageal replacement. This article addresses the progress in esophageal tissue engineering using acellular and cell-seeded approaches. In addition, we discuss the potential direction of future approaches by critically appraising the results in the recent literature.


Scientific Reports | 2016

High contrast microstructural visualization of natural acellular matrices by means of phase-based x-ray tomography

Charlotte K. Hagen; Panagiotis Maghsoudlou; Giorgia Totonelli; Paul C. Diemoz; Marco Endrizzi; Luigi Rigon; Ralf-Hendrik Menk; Fulvia Arfelli; Diego Dreossi; Emmanuel Brun; Paola Coan; Alberto Bravin; Paolo De Coppi; Alessandro Olivo

Acellular scaffolds obtained via decellularization are a key instrument in regenerative medicine both per se and to drive the development of future-generation synthetic scaffolds that could become available off-the-shelf. In this framework, imaging is key to the understanding of the scaffolds’ internal structure as well as their interaction with cells and other organs, including ideally post-implantation. Scaffolds of a wide range of intricate organs (esophagus, lung, liver and small intestine) were imaged with x-ray phase contrast computed tomography (PC-CT). Image quality was sufficiently high to visualize scaffold microarchitecture and to detect major anatomical features, such as the esophageal mucosal-submucosal separation, pulmonary alveoli and intestinal villi. These results are a long-sought step for the field of regenerative medicine; until now, histology and scanning electron microscopy have been the gold standard to study the scaffold structure. However, they are both destructive: hence, they are not suitable for imaging scaffolds prior to transplantation, and have no prospect for post-transplantation use. PC-CT, on the other hand, is non-destructive, 3D and fully quantitative. Importantly, not only do we demonstrate achievement of high image quality at two different synchrotron facilities, but also with commercial x-ray equipment, which makes the method available to any research laboratory.

Collaboration


Dive into the Panagiotis Maghsoudlou's collaboration.

Top Co-Authors

Avatar

Paolo De Coppi

University College London

View shared research outputs
Top Co-Authors

Avatar

Luca Urbani

University College London

View shared research outputs
Top Co-Authors

Avatar

Simon Eaton

University College London

View shared research outputs
Top Co-Authors

Avatar

Giorgia Totonelli

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark Turmaine

University College London

View shared research outputs
Top Co-Authors

Avatar

S Loukogeorgakis

UCL Institute of Child Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alan J. Burns

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge