Network


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

Hotspot


Dive into the research topics where Giuseppe Mazza is active.

Publication


Featured researches published by Giuseppe Mazza.


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.


Epigenetics & Chromatin | 2016

Histone macroH2A1.2 promotes metabolic health and leanness by inhibiting adipogenesis

Valerio Pazienza; Concetta Panebianco; Francesca Rappa; Domenico Memoli; Michela Borghesan; Sara Cannito; Asami Oji; Giuseppe Mazza; Domenico Tamburrino; Giuseppe Fusai; Rosario Barone; Giulia Bolasco; Francesc Villarroya; Joan Villarroya; Kiyotaka Hatsuzawa; Francesco Cappello; Roberta Tarallo; Tomoko M. Nakanishi; Manlio Vinciguerra

BackgroundObesity has tremendous impact on the health systems. Its epigenetic bases are unclear. MacroH2A1 is a variant of histone H2A, present in two alternatively exon-spliced isoforms macroH2A1.1 and macroH2A1.2, regulating cell plasticity and proliferation, during pluripotency and tumorigenesis. Their role in adipose tissue plasticity is unknown.ResultsHere, we show evidence that macroH2A1.1 protein levels in the visceral adipose tissue of obese humans positively correlate with BMI, while macroH2A1.2 is nearly absent. We thus introduced a constitutive GFP-tagged transgene for macroH2A1.2 in mice, and we characterized their metabolic health upon being fed a standard chow diet or a high fat diet. Despite unchanged food intake, these mice exhibit lower adipose mass and improved glucose metabolism both under a chow and an obesogenic diet. In the latter regimen, transgenic mice display smaller pancreatic islets and significantly less inflammation. MacroH2A1.2 overexpression in the mouse adipose tissue induced dramatic changes in the transcript levels of key adipogenic genes; genomic analyses comparing pre-adipocytes to mature adipocytes uncovered only minor changes in macroH2A1.2 genomic distribution upon adipogenic differentiation and suggested differential cooperation with transcription factors. MacroH2A1.2 overexpression markedly inhibited adipogenesis, while overexpression of macroH2A1.1 had opposite effects.ConclusionsMacroH2A1.2 is an unprecedented chromatin component powerfully promoting metabolic health by modulating anti-adipogenic transcriptional networks in the differentiating adipose tissue. Strategies aiming at enhancing macroH2A1.2 expression might counteract excessive adiposity in humans.


PLOS ONE | 2016

Optimization of Liver Decellularization Maintains Extracellular Matrix Micro-Architecture and Composition Predisposing to Effective Cell Seeding

Panagiotis Maghsoudlou; Fanourios Georgiades; Holly Smith; Anna Milan; Panicos Shangaris; Luca Urbani; S Loukogeorgakis; Benedetta Lombardi; Giuseppe Mazza; Charlotte K. Hagen; Nj Sebire; Mark Turmaine; Simon Eaton; Alessandro Olivo; Jasminka Godovac-Zimmermann; Massimo Pinzani; Paul Gissen; Paolo De Coppi

Hepatic tissue engineering using decellularized scaffolds is a potential therapeutic alternative to conventional transplantation. However, scaffolds are usually obtained using decellularization protocols that destroy the extracellular matrix (ECM) and hamper clinical translation. We aim to develop a decellularization technique that reliably maintains hepatic microarchitecture and ECM components. Isolated rat livers were decellularized by detergent-enzymatic technique with (EDTA-DET) or without EDTA (DET). Histology, DNA quantification and proteomics confirmed decellularization with further DNA reduction with the addition of EDTA. Quantification, histology, immunostaining, and proteomics demonstrated preservation of extracellular matrix components in both scaffolds with a higher amount of collagen and glycosaminoglycans in the EDTA-DET scaffold. Scanning electron microscopy and X-ray phase contrast imaging showed microarchitecture preservation, with EDTA-DET scaffolds more tightly packed. DET scaffold seeding with a hepatocellular cell line demonstrated complete repopulation in 14 days, with cells proliferating at that time. Decellularization using DET preserves microarchitecture and extracellular matrix components whilst allowing for cell growth for up to 14 days. Addition of EDTA creates a denser, more compact matrix. Transplantation of the scaffolds and scaling up of the methodology are the next steps for successful hepatic tissue engineering.


Nature Nanotechnology | 2015

Decoupling competing surface binding kinetics and reconfiguration of receptor footprint for ultrasensitive stress assays

Samadhan B. Patil; Manuel Vögtli; Benjamin Webb; Giuseppe Mazza; Massimo Pinzani; Yeong-Ah Soh; Rachel A. McKendry; Joseph W. Ndieyira

Cantilever arrays have been used to monitor biochemical interactions and their associated stress. However, it is often necessary to passivate the underside of the cantilever to prevent unwanted ligand adsorption, and this process requires tedious optimization. Here, we show a way to immobilize membrane receptors on nanomechanical cantilevers so that they can function without passivating the underlying surface. Using equilibrium theory, we quantitatively describe the mechanical responses of vancomycin, human immunodeficiency virus type 1 antigens and coagulation factor VIII captured on the cantilever in the presence of competing stresses from the top and bottom cantilever surfaces. We show that the area per receptor molecule on the cantilever surface influences ligand-receptor binding and plays an important role on stress. Our results offer a new way to sense biomolecules and will aid in the creation of ultrasensitive biosensors.


Amyloid | 2016

Amyloid persistence in decellularized liver: biochemical and histopathological characterization.

Giuseppe Mazza; J. Paul Simons; Raya Al-Shawi; Stephan Ellmerich; Luca Urbani; Sofia Giorgetti; Graham W. Taylor; Janet A. Gilbertson; Andrew R. Hall; W. Al-Akkad; Dipok Kumar Dhar; Philip N. Hawkins; Paolo De Coppi; Massimo Pinzani; Vittorio Bellotti; Palma Mangione

Abstract Systemic amyloidoses are a group of debilitating and often fatal diseases in which fibrillar protein aggregates are deposited in the extracellular spaces of a range of tissues. The molecular basis of amyloid formation and tissue localization is still unclear. Although it is likely that the extracellular matrix (ECM) plays an important role in amyloid deposition, this interaction is largely unexplored, mostly because current analytical approaches may alter the delicate and complicated three-dimensional architecture of both ECM and amyloid. We describe here a decellularization procedure for the amyloidotic mouse liver which allows high-resolution visualization of the interactions between amyloid and the constitutive fibers of the extracellular matrix. The primary structure of the fibrillar proteins remains intact and the amyloid fibrils retain their amyloid enhancing factor activity.


Scientific Reports | 2017

Rapid production of human liver scaffolds for functional tissue engineering by high shear stress oscillation-decellularization

Giuseppe Mazza; W. Al-Akkad; A Telese; L. Longato; Luca Urbani; Benjamin Robinson; Andrew M. Hall; Kenny Kong; L. Frenguelli; Giusi Marrone; Oliver Willacy; Mohsen Shaeri; Alan J. Burns; Massimo Malago; Janet A. Gilbertson; Nigel B. Rendell; Kevin Moore; David J. Hughes; Ioan Notingher; Gavin Jell; Armando del Río Hernández; Paolo De Coppi; K. Rombouts; Massimo Pinzani

The development of human liver scaffolds retaining their 3-dimensional structure and extra-cellular matrix (ECM) composition is essential for the advancement of liver tissue engineering. We report the design and validation of a new methodology for the rapid and accurate production of human acellular liver tissue cubes (ALTCs) using normal liver tissue unsuitable for transplantation. The application of high shear stress is a key methodological determinant accelerating the process of tissue decellularization while maintaining ECM protein composition, 3D-architecture and physico-chemical properties of the native tissue. ALTCs were engineered with human parenchymal and non-parenchymal liver cell lines (HepG2 and LX2 cells, respectively), human umbilical vein endothelial cells (HUVEC), as well as primary human hepatocytes and hepatic stellate cells. Both parenchymal and non-parenchymal liver cells grown in ALTCs exhibited markedly different gene expression when compared to standard 2D cell cultures. Remarkably, HUVEC cells naturally migrated in the ECM scaffold and spontaneously repopulated the lining of decellularized vessels. The metabolic function and protein synthesis of engineered liver scaffolds with human primary hepatocytes reseeded under dynamic conditions were maintained. These results provide a solid basis for the establishment of effective protocols aimed at recreating human liver tissue in vitro.


Journal of Thrombosis and Haemostasis | 2015

Mesenchymal stem cell treatment for hemophilia: a review of current knowledge

Etienne Sokal; Catherine Lombard; Giuseppe Mazza

Hemophilia remains a non‐curative disease, and patients are constrained to undergo repeated injections of clotting factors. In contrast, the sustained production of endogenous factors VIII (FVIII) or IX (FIX) by the patients own cells could represent a curative treatment. Gene therapy has thus provided new hope for these patients. However, the issues surrounding the durability of expression and immune responses against gene transfer vectors remain. Cell therapy, involving stem cells expanded in vitro, can provide de novo protein synthesis and, if implanted successfully, could induce a steady‐state production of low quantities of factors, which may keep the patient above the level required to prevent spontaneous bleeding. Liver‐derived stem cells are already being assessed in clinical trials for inborn errors of metabolism and, in view of their capacity to produce FVIII and FIX in cell culture, they are now also being considered for clinical application in hemophilia patients.


Transplantation | 2017

Biodistribution of Liver-derived Mesenchymal Stem Cells After Peripheral Injection in a Hemophilia A Patient

Etienne Sokal; Catherine Lombard; Véronique Roelants; Mustapha Najimi; Sharat Varma; Camillo Sargiacomo; Joachim Ravau; Giuseppe Mazza; François Jamar; Julia Versavau; Vanessa Jacobs; Marc Jacquemin; Stéphane Eeckhoudt; Catherine Lambert; Xavier Stéphenne; Françoise Smets; Cédric Hermans

Background With the exception of liver transplantation, there is no cure for hemophilia, which is currently managed by preemptive replacement therapy. Liver-derived stem cells are in clinical development for inborn and acquired liver diseases and could represent a curative treatment for hemophilia A. The liver is a major factor VIII (FVIII) synthesis site, and mesenchymal stem cells have been shown to control joint bleeding in animal models of hemophilia. Adult-derived human liver stem cells (ADHLSCs) have mesenchymal characteristics and have been shown able to engraft in and repopulate both animal and human livers. Thus, the objectives were to evaluate the potency of ADHLSCs to control bleeding in a hemophilia A patient and assess the biodistribution of the cells after intravenous injection. Methods A patient suffering from hemophilia A was injected with repeated doses of ADHLSCs via a peripheral vein (35 million 111In-oxine-labeled cells, followed by 125 million cells the next day, and 3 infusions of 250 million cells every 2 weeks thereafter; total infusion period, 50 days). Results After cell therapy, we found a temporary (15 weeks) decrease in the patient’s FVIII requirements and severe bleeding complications, despite a lack of increase in circulating FVIII. The cells were safely administered to the patient via a peripheral vein. Biodistribution analysis revealed an initial temporary entrapment of the cells in the lungs, followed by homing to the liver and to a joint afflicted with hemarthrosis. Conclusion These results suggest the potential use of ADHLSCs in the treatment of hemophilia A.


Advanced Drug Delivery Reviews | 2017

Engineering in vitro models of hepatofibrogenesis

Giuseppe Mazza; W. Al-Akkad; K. Rombouts

ABSTRACT Chronic liver disease is a major cause of morbidity and mortality worldwide marked by chronic inflammation and fibrosis/scarring, resulting in end‐stage liver disease and its complications. Hepatic stellate cells (HSCs) are a dominant contributor to liver fibrosis by producing excessive extracellular matrix (ECM), irrespective of the underlying disease aetiologies, and for many decades research has focused on the development of a number of anti‐fibrotic strategies targeting this cell. Despite major improvements in two‐dimensional systems (2D) by using a variety of cell culture models of different complexity, an efficient anti‐fibrogenic therapy has yet to be developed. The development of well‐defined three‐dimensional (3D) in vitro models, which mimic ECM structures as found in vivo, have demonstrated the importance of cell‐matrix bio‐mechanics, the complex interactions between HSCs and hepatocytes and other non‐parenchymal cells, and this to improve and promote liver cell‐specific functions. Henceforth, refinement of these 3D in vitro models, which reproduce the liver microenvironment, will lead to new objectives and to a possible new era in the search for antifibrogenic compounds. Graphical abstract Figure. No Caption available.


Hepatology Communications | 2018

Liver tissue engineering: From implantable tissue to whole organ engineering

Giuseppe Mazza; W. Al-Akkad; K. Rombouts; Massimo Pinzani

The term “liver tissue engineering” summarizes one of the ultimate goals of modern biotechnology: the possibility of reproducing in total or in part the functions of the liver in order to treat acute or chronic liver disorders and, ultimately, create a fully functional organ to be transplanted or used as an extracorporeal device. All the technical approaches in the area of liver tissue engineering are based on allocating adult hepatocytes or stem cell‐derived hepatocyte‐like cells within a three‐dimensional structure able to ensure their survival and to maintain their functional phenotype. The hosting structure can be a construct in which hepatocytes are embedded in alginate and/or gelatin or are seeded in a pre‐arranged scaffold made with different types of biomaterials. According to a more advanced methodology termed three‐dimensional bioprinting, hepatocytes are mixed with a bio‐ink and the mixture is printed in different forms, such as tissue‐like layers or spheroids. In the last decade, efforts to engineer a cell microenvironment recapitulating the dynamic native extracellular matrix have become increasingly successful, leading to the hope of satisfying the clinical demand for tissue (or organ) repair and replacement within a reasonable timeframe. Indeed, the preclinical work performed in recent years has shown promising results, and the advancement in the biotechnology of bioreactors, ex vivo perfusion machines, and cell expansion systems associated with a better understanding of liver development and the extracellular matrix microenvironment will facilitate and expedite the translation to technical applications. (Hepatology Communications 2018;2:131–141)

Collaboration


Dive into the Giuseppe Mazza's collaboration.

Top Co-Authors

Avatar

Massimo Pinzani

University College London

View shared research outputs
Top Co-Authors

Avatar

K. Rombouts

University College London

View shared research outputs
Top Co-Authors

Avatar

W. Al-Akkad

University College London

View shared research outputs
Top Co-Authors

Avatar

L. Frenguelli

University College London

View shared research outputs
Top Co-Authors

Avatar

A Telese

University College London

View shared research outputs
Top Co-Authors

Avatar

Massimo Malago

University College London

View shared research outputs
Top Co-Authors

Avatar

Kevin Moore

University College London

View shared research outputs
Top Co-Authors

Avatar

Luca Urbani

University College London

View shared research outputs
Top Co-Authors

Avatar

L. Longato

University College London

View shared research outputs
Top Co-Authors

Avatar

Paolo De Coppi

University College London

View shared research outputs
Researchain Logo
Decentralizing Knowledge