Dr Paola Campagnolo


Senior Lecturer in Molecular Cardiovascular Sciences
PhD
+44 (0)1483 684346
32 AY 04

Academic and research departments

School of Biosciences.

About

Biography

Dr Paola Campagnolo is currently Lecturer of Molecular Cardiovascular Biology in the School of Biosciences and Medicine, Faculty of Health and Medical Sciences. Paola graduated in Biotechnology at the University of Padua, Italy. She then moved to Bristol to work under the supervision of Prof Paolo Madeddu at Bristol Heart Institute. In 2009, Paola completed her PhD in Cardiovascular Biology working on the isolation of a patient derived pro-angiogenic progenitor cell population. Next, Paola joined Prof Qingbo Xu's the BHF Centre of Excellence at King's College London as a BHF Research Associate to explore the use of stem cells for the repopulation of vascular grafts. In 2013, Paola joined Prof Molly Stevens' group at the Department of Materials at Imperial College London to undertake the study of novel bio- and nano- materials for cardiovascular tissue engineering.Paola has published extensively in top journals in the field of cardiovascular biology and material science with a total of over 646 citations.Paola is part of the editorial board of Editorial Board of Cardiovascular Biologics and Regenerative Medicine (specialty section of Frontiers in Cardiovascular Medicine), act as a reviewer for several journals, including PLoSONE and ATVB and is part of the reviewing board for the French National Agency of Research and the Italian Ministry of Health.

 

Research collaborations

Within the University of Surrey: Dr Patrizia Camelliti (Department of Biochemical Sciences, FHMS), Dr Vlad Stolojan (FEPS, ATI), Dr Carina Dunlop (FEPS)

External collaborations:

Dr Kevin Maringer (The Pirbright Institute)

Prof Paolo Madeddu (University of Bristol)

Dr Ciro Chiappini (King's College London)

 

Research

Research interests

Teaching

Publications

MARIAM BAPIR, PAOLA CAMPAGNOLO, Ana Rodriguez-Mateos, Simon S. SKENE, CHRISTIAN HEISS (2022)Assessing variability in vascular response to cocoa with personal devices: a series of double-blind randomized cross-over n-of-1 trials, In: Frontiers Nutrition9886597

Controlled clinical intervention studies have demonstrated that cocoa flavanols (CF) can decrease blood pressure and arterial stiffness in healthy humans, although a large variability in the effect size across trials has been reported. Here, we evaluated intra- and inter-individual variability of responses to CF in everyday life using a series of n-of-1 trials in healthy free-living individuals with normal blood pressure carrying personal devices. Eleven healthy young humans participated in a repeated cross-over randomized controlled double-blind n-of-1 trial. On eight consecutive days, each volunteer consumed on alternating days 6 CF capsules (862 mg CF) on four days and 6 matched placebo capsules (P, 0 mg CF/day) on another 4 days in one of two randomized sequences (CF-P-CF-P-CF-P-CF-P or P-CF-P-CF-P-CF-P-CF). On each day the capsules were taken at the same time in the morning with breakfast after baseline measurements. Each subject was provided with an upper arm blood pressure monitor and a finger clip that measures pulse wave velocity (PWV). Measurements of blood pressure, heart rate and PWV were taken at least hourly over 12 hours during the day by the participants. On the first 2 days measurements were performed under supervision to provide training. The overall mixed model analysis showed that CF significantly decreased 12 h systolic blood pressure and PWV by -1.4±0.3 mmHg and - -0.11±0.03 m/s, respectively. Peak effects were observed within the first 3 hours (1.5 h SBP: -4.9±2.2 mmHg, PWV: -0.32±0.17 m/s)and again after 8 h post ingestion. Large inter-individual variation in responses was found (intra-cluster correlation coefficients [ICC]: 0.41, 0.41). When analysing single individuals’ datasets, there was also considerable between-day variation in individual responses that varied greatly between subjects (ICC: 0-0.30, 0-0.22, 0-0.45). Effect sizes inversely correlated with baseline blood pressure values both between-- and within-subjects. The data confirm that cocoa can decrease blood pressure and arterial stiffness in everyday life when elevated within the normal range. The large inter- and intra-individual variation in responses call for more personalized nutritional intervention strategies.

Chloe Jayne Howard, Aumrita Paul, Justin Duruanyanwu, Kenza Sackho, Paola Campagnolo, Vlad Stolojan (2023)The Manufacturing Conditions for the Direct and Reproducible Formation of Electrospun PCL/Gelatine 3D Structures for Tissue Regeneration, In: Nanomaterials (Basel, Switzerland)13(24)3107

Electrospinning is a versatile technique for fabricating nanofibrous scaffolds for tissue engineering applications. However, the direct formation of 3D sponges through electrospinning has previously not been reproducible. We used a Taguchi experimental design approach to optimise the electrospinning parameters for forming PCL and PCL/gelatine 3D sponges. The following parameters were investigated to improve sponge formation: solution concentration, humidity, and solution conductivity. Pure PCL sponges were achievable. However, a much fluffier sponge formed by increasing the solution conductivity with gelatine. The optimal conditions for sponge formation 24 / % 80:20 PCL:gelatine on aluminium foil at ≥70% humidity, 15 cm, 22 kV and 1500 µL/h. The resulting sponge had a highly porous structure with a fibre diameter of ~1 µm. They also supported significantly higher cell viability than 2D electrospun mats, dropcast films of the same material and even the TCP positive control. Our study demonstrates that the direct formation of PCL/gelatine 3D sponges through electrospinning is feasible and promising for tissue engineering applications. The sponges have a highly porous structure and support cell viability, which are essential properties for tissue engineering scaffolds. Further studies are needed to optimise the manufacturing process and evaluate the sponges' long-term performance in vivo.

Mariam Bapir, Gavrielle R. Untracht, Julie E. A. Hunt, John H. McVey, Jenny Harris, Simon S. Skene, Paola Campagnolo, Nikolaos Dikaios, Ana Rodriguez-Mateos, David D Sampson, Danuta M. Sampson, Christian Heiss (2022)Age-Dependent Decline in Common Femoral Artery Flow-Mediated Dilation and Wall Shear Stress in Healthy Subjects, In: Life12(12)2023 MDPI

Femoral artery (FA) endothelial function is a promising biomarker of lower extremity vascular health for peripheral artery disease (PAD) prevention and treatment; however, the impact of age on FA endothelial function has not been reported in healthy adults. Therefore, we evaluated the reproducibility and acceptability of flow-mediated dilation (FMD) in the FA and brachial artery (BA) (n = 20) and performed cross-sectional FA- and BA-FMD measurements in healthy non-smokers aged 22–76 years (n = 50). FMD protocols demonstrated similar good reproducibility. Leg occlusion was deemed more uncomfortable than arm occlusion; thigh occlusion was less tolerated than forearm and calf occlusion. FA-FMD with calf occlusion was lower than BA-FMD (6.0 ± 1.1% vs 6.4 ± 1.3%, p = 0.030). Multivariate linear regression analysis indicated that age (−0.4%/decade) was a significant independent predictor of FA-FMD (R2 = 0.35, p = 0.002). The age-dependent decline in FMD did not significantly differ between FA and BA (pinteraction agexlocation = 0.388). In older participants, 40% of baseline FA wall shear stress (WSS) values were

MARIAM BAPIR, Gabrielle Untracht, Debbie Cooke, John H. McVey, Simon S. SKENE, Paola Campagnolo, Martin B. Whyte, Nikolaos Dikaios, Ana Rodriguez-Mateos, David D Sampson, Danuta M. Sampson, Christian Heiss (2022)Cocoa flavanol consumption improves lower extremity endothelial function in healthy individuals and people with type 2 diabetes, In: Food & Function13(20)pp. 10439-10448 Royal Society of Chemistry

Background: Diabetes and age are major risk factors for the development of lower extremity peripheral artery disease (PAD). Cocoa flavanol (CF) consumption is associated with lower risk for PAD and improves brachial artery (BA) endothelial function. Objectives: to assess if femoral artery (FA) endothelial function and dermal microcirculation are impaired in individuals with type 2 diabetes mellitus (T2DM) and evaluate the acute effect of CF consumption on FA endothelial function. Methods: In a randomised, controlled, double-blind, cross-over study, 22 individuals (n = 11 healthy, n = 11 T2DM) without cardiovascular disease were recruited. Participants received either 1350 mg CF or placebo capsules on 2 separate days in random order. Endothelial function was measured as flow-mediated dilation (FMD) using ultrasound of the common FA and the BA before and 2 hours after interventions. The cutaneous microvasculature was assessed using optical coherence tomography angiography. Results: Baseline FA-FMD and BA-FMD were significantly lower in T2DM (FA: 3.2 ± 1.1% [SD], BA: 4.8 ± 0.8%) compared to healthy (FA: 5.5 ± 0.7%, BA: 6.0 ± 0.8%); each p < 0.001. Whereas in healthy individuals FA-FMD did not significantly differ from BA-FMD (p = 0.144), FA-FMD was significantly lower than BA-FMD in T2DM (p = 0.003) indicating pronounced and additional endothelial dysfunction of lower limb arteries (FA-FMD/BA-FMD: 94 ± 14% [healthy] vs. 68 ± 22% [T2DM], p = 0.007). The baseline FA blood flow rate (0.42 ± 0.23 vs. 0.73 ± 0.35 l min−1, p = 0.037) and microvascular dilation in response to occlusion in hands and feet were significantly lower in T2DM subjects than in healthy ones. CF increased both FA- and BA-FMD at 2 hours, compared to placebo, in both healthy and T2DM subgroups (FA-FMD effect: 2.9 ± 1.4%, BA-FMD effect 3.0 ± 3.5%, each pintervention< 0.001). In parallel, baseline FA blood flow and microvascular diameter significantly increased in feet (3.5 ± 3.5 μm, pintervention< 0.001) but not hands. Systolic blood pressure and pulse wave velocity significantly decreased after CF in both subgroups (−7.2 ± 9.6 mmHg, pintervention = 0.004; −1.3 ± 1.3 m s−1, pintervention = 0.002). Conclusions: Individuals with T2DM exhibit decreased endothelial function that is more pronounced in the femoral than in the brachial artery. CFs increase endothelial function not only in the BA but also the FA both in healthy individuals and in those with T2DM who are at increased risk of developing lower extremity PAD and foot ulcers.

Teemapron Butsabong, Mariana Felippe, Paola Campagnolo, Kevin Maringer (2021)The emerging role of perivascular cells (pericytes) in viral pathogenesis, In: Journal of general virology102(8) Microbiology Soc

Viruses may exploit the cardiovascular system to facilitate transmission or within-host dissemination, and the symptoms of many viral diseases stem at least in part from a loss of vascular integrity. The microvascular architecture is comprised of an endothelial cell barrier ensheathed by perivascular cells (pericytes). Pericytes are antigen-presenting cells (APCs) and play crucial roles in angiogenesis and the maintenance of microvascular integrity through complex reciprocal contact-mediated and paracrine crosstalk with endothelial cells. We here review the emerging ways that viruses interact with pericytes and pay consideration to how these interactions influence microvascular function and viral pathogenesis. Major outcomes of virus-pericyte interactions include vascular leakage or haemorrhage, organ tropism facilitated by barrier disruption, including viral penetration of the blood-brain barrier and placenta, as well as inflammatory, neurological, cognitive and developmental sequelae. The underlying pathogenic mechanisms may include direct infection of pericytes, pericyte modulation by secreted viral gene products and/or the dysregulation of paracrine signalling from or to pericytes. Viruses we cover include the herpesvirus human cytomegalovirus (HCMV, Human betaherpesvirus 5), the retrovirus human immunodeficiency virus (HIV; causative agent of acquired immunodeficiency syndrome, AIDS, and HIV-associated neurocognitive disorder, HAND), the flaviviruses dengue virus (DENV), Japanese encephalitis virus (JEV) and Zika virus (ZIKV), and the coronavirus severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2; causative agent of coronavirus disease 2019, COVID-19). We touch on promising pericyte-focussed therapies for treating the diseases caused by these important human pathogens, many of which are emerging viruses or are causing new or long-standing global pandemics.

VALERIA MASTRULLO, Daan R. van der Veen, ROLANDO SZILVESZTER MATOS, JONATHAN DAVID JOHNSTON, JOHN HENDERSON MCVEY, Paolo Madeddu, EIRINI VELLIOU, PAOLA CAMPAGNOLO, PRIYANKA GUPTA (2022)Pericytes’ Circadian Clock Affects Endothelial Cells’ Synchronization and Angiogenesis in a 3D Tissue Engineered Scaffold, In: Frontiers in pharmacology Frontiers
Davide Maselli, Rolando S. Matos, Robert D. Johnson, Davide Martella, Davide Martella, Valeria Caprettini, Valeria Caprettini, Ciro Chiappini, Ciro Chiappini, Patrizia Camelliti, Paola Campagnolo (2022)Porcine Organotypic Epicardial Slice Protocol: A Tool for the Study of Epicardium in Cardiovascular Research, In: Frontiers in cardiovascular medicine9920013 Frontiers Media S.A

The epicardium has recently gained interest in the cardiovascular field due to its capacity to support heart regeneration after ischemic injury. Models to study the epicardium of large animals in vitro are limited and mainly based on epicardial cell isolation/differentiation from stem cells, followed by 2D cells culture. In this method paper, we describe the procedure to obtain and culture 3D organotypic heart slices presenting an intact epicardium, as a novel model to study the epicardial physiology and activation. Epicardial slices are obtained from porcine hearts using a high-precision vibratome and retain a healthy epicardial layer embedded in its native extracellular environment and connected with other cardiac cells (cardiomyocytes, fibroblasts, vascular cells etc.). Epicardial slices can be cultured for 72 h, providing an ideal model for studying the epicardium physiology or perform pharmacological interventions/gene therapy approaches. We also report on methods to assesses the viability and composition of the epicardial slices, and evaluate their architecture in 3D through tissue decoloration. Finally, we present a potential application for a nanomaterial-based gene transfer method for tracking of epicardial cells within the slice. Crucially, given the similarity in morphology and physiology of porcine heart with its human counterpart, our system provides a platform for translational research while providing a clinically relevant and ethical alternative to the use of small animals in this type of research.

Paola Campagnolo, Maurizio Pesce (2021)Editorial: Bio-materials for Cardiovascular Diseases, In: Frontiers in Cardiovascular Medicine8 Frontiers Media S.A
Yin P Cheung, Valeria Mastrullo, Davide Maselli, Teemapron Butsabong, Paolo Madeddu, Kevin Maringer, Paola Campagnolo (2020)A Critical Role for Perivascular Cells in Amplifying Vascular Leakage Induced by Dengue Virus Nonstructural Protein 1, In: mSphere5(4)e00258-20pp. 1-11

Dengue is the most prevalent arthropod-borne viral disease affecting humans, with severe dengue typified by potentially fatal microvascular leakage and hypovolemic shock. Blood vessels of the microvasculature are composed of a tubular structure of endothelial cells ensheathed by perivascular cells (pericytes). Pericytes support endothelial cell barrier formation and maintenance through paracrine and contact-mediated signaling and are critical to microvascular integrity. Pericyte dysfunction has been linked to vascular leakage in noncommunicable pathologies such as diabetic retinopathy but has never been linked to infection-related vascular leakage. Dengue vascular leakage has been shown to result in part from the direct action of the secreted dengue virus (DENV) nonstructural protein NS1 on endothelial cells. Using primary human vascular cells, we show here that NS1 also causes pericyte dysfunction and that NS1-induced endothelial hyperpermeability is more pronounced in the presence of pericytes. Notably, NS1 specifically disrupted the ability of pericytes to support endothelial cell function in a three-dimensional (3D) microvascular assay, with no effect on pericyte viability or physiology. These effects are mediated at least in part through contact-independent paracrine signals involved in endothelial barrier maintenance by pericytes. We therefore identify a role for pericytes in amplifying NS1-induced microvascular hyperpermeability in severe dengue and thus show that pericytes can play a critical role in the etiology of an infectious vascular leakage syndrome. These findings open new avenues of research for the development of drugs and diagnostic assays for combating infection-induced vascular leakage, such as severe dengue. The World Health Organization considers dengue one of the top 10 global public health problems. There is no specific antiviral therapy to treat dengue virus and no way of predicting which patients will develop potentially fatal severe dengue, typified by vascular leakage and circulatory shock. We show here that perivascular cells (pericytes) amplify the vascular leakage-inducing effects of the dengue viral protein NS1 through contact-independent signaling to endothelial cells. While pericytes are known to contribute to noncommunicable vascular leakage, this is the first time these cells have been implicated in the vascular effects of an infectious disease. Our findings could pave the way for new therapies and diagnostics to combat dengue and potentially other infectious vascular leakage syndromes.

Nelson Chong, Paola Campagnolo (2023)Editorial: Circadian biology, cardiovascular function and disease, In: Frontiers in pharmacology14 Frontiers Media Sa
D. Maselli, R. S. Matos, R. D. Johnson, C. Chiappini, P. Camelliti, P. Campagnolo (2022)Epicardial slices: an innovative 3D organotypic model to study epicardial cell physiology and activation, In: npj Regenerative medicine7(1)pp. 7-7 Nature Publishing Group UK

The epicardium constitutes an untapped reservoir for cardiac regeneration. Upon heart injury, the adult epicardium re-activates, leading to epithelial-to-mesenchymal transition (EMT), migration, and differentiation. While interesting mechanistic and therapeutic findings arose from lower vertebrates and rodent models, the introduction of an experimental system representative of large mammals would undoubtedly facilitate translational advancements. Here, we apply innovative protocols to obtain living 3D organotypic epicardial slices from porcine hearts, encompassing the epicardial/myocardial interface. In culture, our slices preserve the in vivo architecture and functionality, presenting a continuous epicardium overlaying a healthy and connected myocardium. Upon thymosin β4 treatment of the slices, the epicardial cells become activated, upregulating epicardial and EMT genes, resulting in epicardial cell mobilization and differentiation into epicardial-derived mesenchymal cells. Our 3D organotypic model enables to investigate the reparative potential of the adult epicardium, offering an advanced tool to explore ex vivo the complex 3D interactions occurring within the native heart environment.

Daniella A Lefteri, Steven R Bryden, Marieke Pingen, Sandra Terry, Ailish McCafferty, Emily F Beswick, Georgi Georgiev, Marleen Van der Laan, Valeria Mastrullo, Paola Campagnolo, Robert M Waterhouse, Margus Varjak, Andres Merits, Rennos Fragkoudis, Stephen Griffin, Kave Shams, Emilie Pondeville, Clive S McKimmie (2022)Mosquito saliva enhances virus infection through sialokinin-dependent vascular leakage, In: Proceedings of the National Academy of Sciences - PNAS119(24)pp. e2114309119-e2114309119

Viruses transmitted by mosquitoes are an increasingly important global cause of disease. Defining common determinants of host susceptibility to this large group of heterogenous pathogens is key for informing the rational design of panviral medicines. Infection of the vertebrate host with these viruses is enhanced by mosquito saliva, a complex mixture of salivary-gland-derived factors and microbiota. We show that the enhancement of infection by saliva was dependent on vascular function and was independent of most antisaliva immune responses, including salivary microbiota. Instead, the gene product sialokinin mediated the enhancement of virus infection through a rapid reduction in endothelial barrier integrity. Sialokinin is unique within the insect world as having a vertebrate-like tachykinin sequence and is absent from , which are incompetent for most arthropod-borne viruses, whose saliva was not proviral and did not induce similar vascular permeability. Therapeutic strategies targeting sialokinin have the potential to limit disease severity following infection with -mosquito-borne viruses.

Ciro Chiappini, Enrica De Rosa, Jonathan Otto Martinez, Paola Campagnolo, Carina Almeida, Ennio Tasciotti, Molly Stevens (2015)Porous Silicon Nanoneedles By Metal Assisted Chemical Etch for Intracellular Sensing and Delivery, In: ECS transactions69(2)pp. 63-68

Metal assisted chemical etch has recently come to prominence as a versatile strategy for the realization of silicon nanostructures with tailored porosity. By exploiting metal assisted chemical etch, we recently developed porous silicon nanoneedles capable of interfacing with cells for delivery to and sensing of the intracellular milieu. Here we review our recently published studies on the fabrication of such nanostructures. Further we review their use as vectors for the localized delivery nucleic acids capable of inducing neovasculature formation in a mouse model. Finally we provide an overview of our findings on the use of porous silicon nanoneedles as intracellular sensors for detection of enzymatic activity with high resolution across excised human tissue samples.

DAVIDE MASELLI, ROLANDO SZILVESZTER MATOS, ROBERT DAVID JOHNSON, C Chiappini, PATRIZIA CAMELLITI, PAOLA CAMPAGNOLO (2021)Epicardial slices: a 3D organotypic model for the study of epicardial activation and differentiation, In: NPJ Regenerative medicine Springer

The epicardium constitutes an untapped reservoir for cardiac regeneration. Upon heart injury, the adult epicardium re-activates, leading to epithelial-to-mesenchymal transition (EMT), migration and differentiation. While interesting mechanistic and therapeutic findings arose from lower vertebrates and rodent models, the introduction of an experimental system representative of large mammals would undoubtedly facilitate translational advancements. Here, we apply innovative protocols to obtain living 3D organotypic epicardial slices from porcine hearts, encompassing the epicardial/myocardial interface. In culture, our slices preserve the in vivo architecture and functionality, presenting a continuous epicardium overlaying a healthy and connected myocardium. Upon thymosin β4 treatment of the slices, the epicardial cells become activated upregulating epicardial and EMT genes, resulting in epicardial cell mobilization and differentiation into epicardial-derived mesenchymal cells. Our 3D organotypic model enables to investigate the reparative potential of the adult epicardium, offering an advanced tool to explore ex vivo the complex 3D interactions occurring within the native heart environment

Yin P. Cheung, Valeria Mastrullo, Davide Maselli, Teemapron Butsabong, Paolo Madeddu, Kevin Maringer, Paola Campagnolo (2020)A Critical Role for Perivascular Cells in Amplifying Vascular Leakage Induced by Dengue Virus Non-Structural Protein 1, In: mSphere American Society for Microbiology

Dengue is the most prevalent arthropod-borne viral disease affecting humans, with severe dengue typified by potentially fatal microvascular leakage and hypovolaemic shock. Blood vessels of the microvasculature are composed of a tubular structure of endothelial cells ensheathed by perivascular cells (pericytes). Pericytes support endothelial cell barrier formation and maintenance through paracrine and contact-mediated signalling, and are critical to microvascular integrity. Pericyte dysfunction has been linked to vascular leakage in noncommunicable pathologies such as diabetic retinopathy, but has never been linked to infection-related vascular leakage. Dengue vascular leakage has been shown to result in part from the direct action of the secreted dengue virus (DENV) non-structural protein NS1 on endothelial cells. Using primary human vascular cells, we show here that NS1 also causes pericyte dysfunction, and that NS1-induced endothelial hyperpermeability is more pronounced in the presence of pericytes. Notably, NS1 specifically disrupted the ability of pericytes to support endothelial cell function in a 3D microvascular assay, with no effect on pericyte viability or physiology. These effects are mediated at least in part through contact-independent paracrine signals involved in endothelial barrier maintenance by pericytes. We therefore identify a role for pericytes in amplifying NS1-induced microvascular hyperpermeability in severe dengue, and thus show that pericytes can play a critical role in the aetiology of an infectious vascular leakage syndrome. These findings open new avenues of research for the development of drugs and diagnostic assays for combating infection-induced vascular leakage, such as severe dengue.

Elisa Avolio, Paola Campagnolo, Rajesh Katare, Paolo Madeddu (2023)The role of cardiac pericytes in health and disease: therapeutic targets for myocardial infarction, In: Nature reviews cardiology Nature Research

Millions of cardiomyocytes die immediately after myocardial infarction, regardless of whether the culprit coronary artery undergoes prompt revascularization. Residual ischaemia in the peri-infarct border zone causes further cardiomyocyte damage, resulting in a progressive decline in contractile function. To date, no treatment has succeeded in increasing the vascularization of the infarcted heart. In the past decade, new approaches that can target the heart's highly plastic perivascular niche have been proposed. The perivascular environment is populated by mesenchymal progenitor cells, fibroblasts, myofibroblasts and pericytes, which can together mount a healing response to the ischaemic damage. In the infarcted heart, pericytes have crucial roles in angiogenesis, scar formation and stabilization, and control of the inflammatory response. Persistent ischaemia and accrual of age-related risk factors can lead to pericyte depletion and dysfunction. In this Review, we describe the phenotypic changes that characterize the response of cardiac pericytes to ischaemia and the potential of pericyte-based therapy for restoring the perivascular niche after myocardial infarction. Pericyte-related therapies that can salvage the area at risk of an ischaemic injury include exogenously administered pericytes, pericyte-derived exosomes, pericyte-engineered biomaterials, and pharmacological approaches that can stimulate the differentiation of constitutively resident pericytes towards an arteriogenic phenotype. Promising preclinical results from in vitro and in vivo studies indicate that pericytes have crucial roles in the treatment of coronary artery disease and the prevention of post-ischaemic heart failure.

Rachel E. Smith, Stella Totti, Eirini Velliou, Paola Campagnolo, Suzanne M. Hingley-Wilson, Neil I. Ward, John R. Varcoe, Carol Crean (2019)Development of a novel highly conductive and flexible cotton yarn for wearable pH sensor technology, In: Sensors and Actuators B: Chemical287pp. 338-345 Elsevier

The simple and effective approach of “dipping and drying” cotton yarn in a dispersion of poly(3,4-ethylenedioxythiophene)-poly(styrene sulfonate) (PEDOT:PSS) and multi-walled carbon nanotubes (MWCNT) resulted in the development of a highly conductive and flexible cotton fibres. Subsequent polyaniline (PANi) deposition yielded electrodes with significant biocompatible and antibacterial properties that could be fabricated (alongside quasi-reference electrodes) into solid-state wearable pH sensors, which achieve rapid, selective, and Nernstian responses (-61 ± 2 mV pH-1) over a wide pH range (2.0 – 12.0), even in a pH-adjusted artificial sweat matrix. This development represents an important progression towards the realisation of real-time, on-body, wearable sensors.

P Campagnolo, AJ Gormley, LW Chow, AG Guex, PA Parmar, JL Puetzer, JAM Steele, A Breant, P Madeddu, MM Stevens (2016)Pericyte seeded dual peptide scaffold with improved endothelialization for vascular graft tissue engineering, In: Advanced healthcare materials

The development of synthetic vascular grafts for coronary artery bypass is challenged by insufficient endothelialization, which exposes to the risk of thrombosis, and lack of native cellular constituents, which favours pathological remodelling. Here, an bifunctional electrospun poly(ε-caprolactone) (PCL) scaffold with potential for synthetic vascular graft applications is presented. This scaffold incorporates two tethered peptides: the osteopontin-derived peptide (Adh) on the ‘luminal’ side and a heparin-binding peptide (Hep) on the ‘abluminal’ side. Additionally, the ‘abluminal’ side of the scaffold is seeded with saphenous vein-derived pericytes (SVPs) as a source of pro-angiogenic growth factors. The Adh peptide significantly increase endothelial cell adhesion, while the Hep peptide promote accumulation of vascular endothelial growth factor (VEGF) secreted by SVPs. SVPs increase endothelial migration both in a transwell assay and a modified scratch assay performed on the PCL scaffold. Seeding of SVPs on the ‘abluminal’/Hep side of the scaffold further increase endothelial cell density, indicating a combinatory effect of the peptides and pericytes. Lastly, SVP-seeded scaffolds are preserved by freezing in a xeno-free medium, maintaining good cell viability and function. In conclusion, this engineered scaffold combines patient-derived pericytes and spatially organized functionalities, which synergistically increase endothelial cell density and growth factor retention.

E Avolio, VV Alvino, MT Ghorbel, Paola Campagnolo (2016)Perivascular cells and tissue engineering: current applications and untapped potential, In: Pharmacology and Therapeutics171pp. 83-92 Elsevier

The recent development of tissue engineering provides exciting new perspectives for the replacement of failing organs and the repair of damaged tissues. Perivascular cells, including vascular smooth muscle cells, pericytes and other tissue specific populations residing around blood vessels, have been isolated from many organs and are known to participate to the in situ repair process and angiogenesis. Their potential has been harnessed for cell therapy of numerous pathologies; however, in this Review we will discuss the potential of perivascular cells in the development of tissue engineering solutions for healthcare. We will examine their application in the engineering of vascular grafts, cardiac patches and bone substitutes as well as other tissue engineering applications and we will focus on their extensive use in the vascularization of engineered constructs. Additionally, we will discuss the emerging potential of human pericytes for the development of efficient, vascularized and non-immunogenic engineered constructs.

G Luongo, Paola Campagnolo, JE Perez, J Kosel, TK Georgiou, A Regoutz, DJ Payne, MM Stevens, MP Ryan, AE Porter, IE Dunlop (2017)Scalable high-affinity stabilization of magnetic iron oxide nanostructures by a biocompatible antifouling homopolymer, In: ACS Applied Materials & Interfaces9(46)pp. 40059-40069 American Chemical Society

Iron oxide nanostructures have been widely developed for biomedical applications, due to their magnetic properties and biocompatibility. In clinical application, the stabilization of these nanostructures against aggregation and non-specific interactions is typically achieved using weakly anchored polysaccharides, with better-defined and more strongly anchored synthetic polymers not commercially adopted due to complexity of synthesis and use. Here, we show for the first time stabilization and biocompatibilization of iron oxide nanoparticles by a synthetic homopolymer with strong surface anchoring and a history of clinical use in other applications, poly(2-methacryloyloxyethy phosphorylcholine) (poly(MPC)). For the commercially important case of spherical particles, binding of poly(MPC) to iron oxide surfaces and highly effective individualization of magnetite nanoparticles (20 nm) are demonstrated. Next-generation highaspect ratio nanowires (both magnetite/maghemite and core-shell iron/iron oxide) are furthermore stabilized by poly(MPC)-coating, with nanowire cytotoxicity at large concentrations significantly reduced. The synthesis approach is exploited to incorporate functionality into the poly(MPC) chain is demonstrated by random copolymerization with an alkyne-containing monomer for click-chemistry. Taking these results together, poly(MPC) homopolymers and random copolymers offer a significant improvement over current iron oxide nanoformulations, combining straightforward synthesis, strong surface-anchoring and well-defined molecular weight.

Valeria Mastrullo, William Cathery, Eirini Velliou, Paolo Madeddu, Paola Campagnolo (2020)Angiogenesis in Tissue Engineering: As Nature Intended?, In: Ranieri Cancedda (eds.), Frontiers in Bioengineering and Biotechnology Frontiers Media

Despite the steady increase in the number of studies focusing on the development of tissue engineered constructs, solutions delivered to the clinic are still limited. Specifically, the lack of mature and functional vasculature greatly limits the size and complexity of vascular scaffold models. If tissue engineering aims to replace large portions of tissue with the intention of repairing significant defects, a more thorough understanding of the mechanisms and players regulating the angiogenic process is required in the field. This review will present the current material and technological advancements addressing the imperfect formation of mature blood vessels within tissue engineered structures.

E Di Bernardini, P Campagnolo, A Margariti, A Zampetaki, E Karamariti, Y Hu, Q Xu (2014)Endothelial lineage differentiation from induced pluripotent stem cells is regulated by microRNA-21 and transforming growth factor β2 (TGF-β2) pathways., In: The Journal of Biological Chemistry289(6)pp. 3383-3393 The American Society for Biochemistry and Molecular Biology, Inc.

Finding a suitable cell source for endothelial cells (ECs) for cardiovascular regeneration is a challenging issue for regenerative medicine. In this paper, we describe a novel mechanism regulating induced pluripotent stem cells (iPSC) differentiation into ECs, with a particular focus on miRNAs and their targets. We first established a protocol using collagen IV and VEGF to drive the functional differentiation of iPSCs into ECs and compared the miRNA signature of differentiated and undifferentiated cells. Among the miRNAs overrepresented in differentiated cells, we focused on microRNA-21 (miR-21) and studied its role in iPSC differentiation. Overexpression of miR-21 in predifferentiated iPSCs induced EC marker up-regulation and in vitro and in vivo capillary formation; accordingly, inhibition of miR-21 produced the opposite effects. Importantly, miR-21 overexpression increased TGF-2 mRNA and secreted protein level, consistent with the strong up-regulation of TGF-2 during iPSC differentiation. Indeed, treatment of iPSCs with TGF-2 induced EC marker expression and in vitro tube formation. Inhibition of SMAD3, a downstream effector of TGF-2, strongly decreased VE-cadherin expression. Furthermore,TGF-2 neutralization and knockdown inhibited miR-21-induced EC marker expression. Finally, we confirmed the PTEN/Akt pathway as a direct target of miR-21, and we showed that PTEN knockdown is required for miR-21-mediated endothelial differentiation. In conclusion, we elucidated a novel signaling pathway that promotes the differentiation of iPSC into functional ECs suitable for regenerative medicine applications.

M Gubernator, SC Slater, HL Spencer, I Spiteri, A Sottoriva, F Riu, J Rowlinson, E Avolio, R Katare, G Mangialardi, A Oikawa, C Reni, P Campagnolo, G Spinetti, A Touloumis, S Tavare, F Prandi, M Pesce, M Hofner, V Klemens, C Emanueli, G Angelini, P Madeddu (2015)Epigenetic Profile of Human Adventitial Progenitor Cells Correlates With Therapeutic Outcomes in a Mouse Model of Limb Ischemia, In: ARTERIOSCLEROSIS THROMBOSIS AND VASCULAR BIOLOGY35(3)pp. 675-688 LIPPINCOTT WILLIAMS & WILKINS
LS Barcelos, C Duplaa, N Kraenkel, G Graiani, G Invernici, R Katare, M Siragusa, M Meloni, I Campesi, M Monica, A Simm, P Campagnolo, G Mangialardi, L Stevanato, G Alessandri, C Emanueli, P Madeddu (2009)Human CD133(+) Progenitor Cells Promote the Healing of Diabetic Ischemic Ulcers by Paracrine Stimulation of Angiogenesis and Activation of Wnt Signaling, In: CIRCULATION RESEARCH104(9)pp. 1095-U199 LIPPINCOTT WILLIAMS & WILKINS
Mona N. Sweilam, Sarah F. Cordery, Stella Totti, Eirini G. Velliou, Paola Campagnolo, John R. Varcoe, M. Begoña Delgado-Charro, Carol Crean (2020)Textile-based non-invasive lithium drug monitoring: a proof-of-concept study for wearable sensing, In: Biosensors and Bioelectronics150111897 Elsevier

Flexible wearable chemical sensors are emerging tools which target diagnosis and monitoring of medical conditions. One of the potential applications of wearable chemical sensors is therapeutic drug monitoring for drugs that have a narrow therapeutic range such as lithium. We have investigated the possibility of developing a fibre-based device for non-invasive lithium drug monitoring in interstitial fluid. A flexible cotton-based lithium sensor was coupled with a carbon fibre-based reference electrode to obtain a potentiometric device. In vitro reverse iontophoresis experiments were performed to extract Li+ from under porcine skin by applying a current density of 0.4 mA cm-2 via two electrodes. Carbon fibre-based reverse iontophoresis electrodes were fabricated and used instead of a conventional silver wire-based version and comparable results were obtained. The fibre-based Li+ sensor and reference electrodes were capable of determining the Li+ concentration in samples collected via reverse iontophoresis and the results compared well to those obtained by ion chromatography. Additionally, biocompatibility of the used materials have been tested. Promising results were obtained which confirm the possibility of monitoring lithium in interstitial fluid using a wearable sensor.

Rolando S. Matos, Davide Maselli, Christian Heiss, Paola Campagnolo, John Henderson Mcvey (2022)3D printed bioreactor enabling the pulsatile culture of native and angioplastied large arteries, In: Frontiers in Cardiovascular Medicine9864580 Frontiers Media

Routine interventions such as balloon angioplasty, result in vascular activation and remodeling, often requiring re-intervention. 2D in vitro models and small animal experiments have enabled the discovery of important mechanisms involved in this process, however the clinical translation is often underwhelming. There is a critical need for an ex vivo model representative of the human vascular physiology and encompassing the complexity of the vascular wall and the physical forces regulating its function. Vascular bioreactors for ex vivo culture of large vessels are viable alternatives, but their custom-made design and insufficient characterization often hinders the reproducibility of the experiments. The objective of the study was to design and validate a novel 3D printed cost-efficient and versatile perfusion system, capable of sustaining the viability and functionality of large porcine arteries for 7 days and enabling early post-injury evaluations. MultiJet Fusion 3D printing was used to engineer the EasyFlow insert, converting a conventional 50 ml centrifuge tube into a mini bioreactor. Porcine carotid arteries either left untreated or injured with an angioplasty balloon, were cultured under pulsatile flow for up to 7 days. Pressure, heart rate, medium viscosity and shear conditions were adjusted to resemble arterial in vivo hemodynamics. Tissue viability, cell activation and matrix remodeling were analyzed by immunohistochemistry, and vascular function was monitored by duplex ultrasound. Culture conditions in the EasyFlow bioreactor preserved endothelial coverage and smooth muscle organization and extracellular matrix structure in the vessel wall, as compared to static culture. Injured arteries presented hallmarks of early remodeling, such as intimal denudation, smooth muscle cell disarray and media/adventitia activation in flow culture. Duplex ultrasound confirmed continuous pulsatile blood flow conditions, dose-dependent vasodilator response to nitroglycerin in untreated vessels and impaired dilator response in angioplastied vessels. The scope of this work is to validate a low-cost, robust and reproducible system to explore the culture of native and injured large arteries under pulsatile flow. While the study of vascular pathology is beyond the scope of the present paper, our system enables future investigations and provides a platform to test novel therapies and devices ex vivo, in a patient relevant system.

Background Vessel graft failure is typically associated neointimal formation and arteriosclerosis, in which endothelial dysfunction/damage is a key event.1 Resveratrol has been shown to possess cardioprotective capacity and to reduce atherosclerosis, through its anti-oxidant and anti-apoptotic properties.2–4 However, it is unknown whether it influences the behavior of resident stem cells in the vessel wall leading to the development of arteriosclerosis. Methods and results In the present study, the mouse model of vein grafts was established by grafting vena cava to carotid artery using a cuff technique. ApoE knockout animals were randomly assigned to two groups, one receiving a diet enriched with resveratrol and one control group on a normal diet. It was found that neointimal lesions 4 weeks postoperatively were significantly reduced in resveratrol-treated group compared to untreated controls. Immunostaining for cell components in the grafts revealed the presence of Sca-1+ progenitor cells in the lesional adventitia and neointima of vein grafts. Subsequently, Sca-1+ cells from 4-week vein grafts were cultivated and isolated. Interestingly, stem/progenitor cell differentiation into endothelial lineage was markedly increased by treatment with resveratrol in vitro. We investigated the mechanism involved in the resveratrol-induced progenitor cell differentiation and identified miR-21 as a target of resveratrol. We demonstrated that resveratrol significantly reduced miR-21 expression during endothelial differentiation of progenitor cells, which in turn reduced Akt phosphorylation. This signal cascade diminished the amount of nuclear β-catenin, ultimately inducing endothelial marker expression and tube-like formation capacity in stem/progenitor cells. Both the inhibition of miR-21 and the knockdown of β-catenin were able to resemble the effect of resveratrol application, i.e. reducing endothelial differentiation. Finally, the effect of resveratrol on progenitor cell differentiation was blunted by the overexpression of miR-21. Conclusion We provide the first evidence that oral administration of resveratrol can reduce neointimal formation in an animal model of vein graft, by inducing re-endothelialization through progenitor cell differentiation. We established that the mechanism involved is miR-21/Akt/β-catenin dependent. These findings might contribute to explain the beneficial effect of red wine consumption on vascular disease.

P Campagnolo, J P Kirton, T Tsai, E Di Bernardini, Q Xu (2011)50 Embryonic and IPS cell-derived cKit+ cells represent a novel vascular progenitor cell population, In: Heart (British Cardiac Society)97(20)pp. e7-e7 BMJ Publishing Group Ltd and British Cardiovascular Society

The therapeutic exploitation of stem cell-derived vascular progenitor cells is limited by the heterogeneity of the cultures and by an incomplete understanding of molecular switches that control their differentiation either to endothelial or smooth muscle cells. We investigated the vascular potential of c-Kit+ cells isolated from mouse embryonic and induced pluripotent stem cells and identified a novel mechanism regulating their differentiation. In the present study, mouse embryonic stem cells were cultured on collagen IV-coated plates for 4 days and then the mixed population containing about 50% c-Kit+ cells was sorted. c-Kit+ progenitor cells were stimulated with PDGF or VEGF. Culture with PDGF gave rise to α-smooth muscle actin, calponin and smooth muscle myosin heavy chain positive smooth muscle cells. In the presence of VEGF, c-Kit+ cells differentiate towards endothelial lineage expressing Flk1, CD31, VE-cadherin and vWF. In particular, exposure to shear stress between day 3 and 5 significantly increased the expression of endothelial markers and improved tube formation capacity as shown in a 3-dimensional matrix assay. High level of Klf4 was detected at early stage of endothelial differentiation while a strong reduction occurred during smooth muscle differentiation. Indeed, infection with Klf4 adenovirus strongly induced differentiation to endothelial cells while strongly repressed smooth muscle markers. Thus, our data demonstrated for the first time that c-Kit+ cells are a suitable source of vascular progenitor cells and their differentiation is regulated by a key transcription factor, namely Klf4.

Nicolle Kränkel, Rajesh Katare, Mauro Siragusa, Luciola Barcelos, Paola Campagnolo, Giuseppe Mangialardi, Orazio Fortunato, Gaia Spinetti, Nguyen Tran, Kai Zacharowski, Wojciech Wojakowski, Iwona Mroz, Andrew Herman, Jocelyn Manning Fox, Patrick Macdonald, Joost Schanstra, Jean-Loup Bascands, Raimondo Ascione, Gianni Angelini, Costanza Emanueli, Paolo Madeddu (2008)Role of kinin B2 receptor signaling in the recruitment of circulating progenitor cells with neovascularization potential, In: Circulation research103(11)pp. 1335-43 American Heart Association

Reduced migratory function of circulating angiogenic progenitor cells (CPCs) has been associated with impaired neovascularization in patients with cardiovascular disease (CVD). Previous findings underline the role of the kallikrein-kinin system in angiogenesis. We now demonstrate the involvement of the kinin B2 receptor (B(2)R) in the recruitment of CPCs to sites of ischemia and in their proangiogenic action. In healthy subjects, B(2)R was abundantly present on CD133(+) and CD34(+) CPCs as well as cultured endothelial progenitor cells (EPCs) derived from blood mononuclear cells (MNCs), whereas kinin B1 receptor expression was barely detectable. In transwell migration assays, bradykinin (BK) exerts a potent chemoattractant activity on CD133(+) and CD34(+) CPCs and EPCs via a B(2)R/phosphoinositide 3-kinase/eNOS-mediated mechanism. Migration toward BK was able to attract an MNC subpopulation enriched in CPCs with in vitro proangiogenic activity, as assessed by Matrigel assay. CPCs from cardiovascular disease patients showed low B(2)R levels and decreased migratory capacity toward BK. When injected systemically into wild-type mice with unilateral limb ischemia, bone marrow MNCs from syngenic B(2)R-deficient mice resulted in reduced homing of sca-1(+) and cKit(+)flk1(+) progenitors to ischemic muscles, impaired reparative neovascularization, and delayed perfusion recovery as compared with wild-type MNCs. Similarly, blockade of the B(2)R by systemic administration of icatibant prevented the beneficial effect of bone marrow MNC transplantation. BK-induced migration represents a novel mechanism mediating homing of circulating angiogenic progenitors. Reduction of BK sensitivity in progenitor cells from cardiovascular disease patients might contribute to impaired neovascularization after ischemic complications.

P Campagnolo, A J Gormley, L W Chow, A G Guex, P A Parmar, J L Puetzer, JAM Steele, P Madeddu, M M Stevens (2016)P36 Pericyte seeded biofunctionalised vascular graft for improved endothelial coverage, In: Heart (British Cardiac Society)102(Suppl 8)

Coronary bypass is the most common intervention for coronary obstruction. When autologous vessels are unavailable, synthetic grafts are employed. The challenge of rapid endothelialisation to reduce complications is still far from being achieved. This study describes a novel approach to synthetic graft design, based on the spatially defined presentation of functionalities within the scaffold, contributing to the biological activity of the pre-seeded pro-angiogenic human saphenous vein-derived pericytes (SVP). Indeed, the electrospun polycaprolactone (PCL) presented two distinct functionalities on either side: the osteopontin-derived adhesion peptide (Adh) to facilitate endothelial adhesion on the luminal side and the heparin binding peptide (Hep) to create a reservoir of SVP-produced growth factors on the adventitial side. Conjugated peptides retained functionality within the scaffold, with Adh increasing endothelial cells (EC) adhesion and Hep binding the SVP-produced VEGF and releasing it to improve EC growth over time. Additionally, seeding of the scaffold with SVP increased EC migration and proliferation, suggesting their supporting role in re-endothelialisation of the graft. When seeded on the dual peptide scaffold, SVP acted synergistically with the peptides determining a greater EC coverage, as compared to each component alone. SVP-seeded scaffolds were cryopreserved for up to 2 weeks in GMP-grade medium preserving cell viability and functionality. In conclusion, we engineered a progenitor cell-seeded vascular graft, which incorporates spatially organised biomolecules that act synergistically with the cells to support EC coverage.

E Di Bernardini, P Campagnolo, A Margariti, A Zampetaki, M Mayer, Y Hu, Q Xu (2013)183 ENDOTHELIAL LINEAGE DIFFERENTIATION FROM IPS CELLS IS REGULATED BY MIRNA-21/AKT AND TGF-[Beta]2 PATHWAYS, In: Heart (British Cardiac Society)99(suppl 2) BMJ Publishing Group LTD

Background Endothelial death/dysfunction is a critical process in the development of cardiovascular diseases. Finding a source of endothelial cells (ECs) for regenerative medicine is a challenging yet fundamental issue. Induced pluripotent stem cells (iPSCs) constitute an attractive source of cells for transplantation because of their high proliferation and differentiation potential. Methods and Results In this study, we investigated the mechanisms regulating iPSC differentiation into ECs, focusing on miRNAs and their targets. Firstly, we established a differentiation protocol using collagen and VEGF to drive the functional differentiation of iPSCs into ECs. After 7 days, the cells strongly expressed EC markers such as VE-cadherin, Flk-1, vWF and eNOS and formed tubes on Matrigel culture. A parallel study was conducted on embryonic stem cells (ESCs) and iPSCs to analyze the synergistic effect of VEGF and shear stress in improving EC differentiation. Next, we compared the miRNA signature of undifferentiated and differentiated cells with VEGF for 3 days. Amongst the 5 validated miRNAs we focused on miR-21, previously shown to be involved in angiogenesis. Overexpression of miR-21 (pre-21) in pre-differentiated iPSCs induced EC marker upregulation, while inhibition miR-21 (LNA-21) produced the opposite effect. Accordingly, in vitro and in vivo capillary formation was increased by pre-21 and reduced by LNA-21. Interestingly, miR-21 overexpression increased TGFβ2 mRNA and secreted protein level, which was consistent with the strong upregulation of TGFβ-2 during VEGF-induced iPSC differentiation. In addition treatment of iPSCs with TGFβ-2 induced EC marker expression and in vitro tube formation through induction of VEGF secretion. Inhibition of SMAD3, a downstream effector of TGFβ-2, strongly decreased VE-cadherin expression, indicating the importance of the TGFβ-2 pathway in the differentiation process. Furthermore, TGFβ-2 neutralizing antibody inhibited miR-21 induced EC marker upregulation, indicating that TGFβ-2 is a required downstream effecter for miR-21-mediated EC differentiation. In silico target screening revealed PTEN as one of the predicted targets of miR-21. Our results confirmed that pre-21 significantly inhibited PTEN expression whereas LNA-21 induced it. Luciferase assay confirmed that miR-21directly binds to the 3' UTR of PTEN, which is inhibited by a site-specific mutation. Since PTEN is the upstream negative regulator of AKT signaling pathway, we then confirmed that pre-21 induced Akt phosphorylation, while LNA-21 decreased it. Functionally, it was found that inhibition of miR-21 in vivo Matrigel assay revealed the significant influence on angiogenesis in animal models. Conclusion These data demonstrate that miR-21 directly targets the PTEN/Akt pathway, in which TGFβ-2 pathway regulated by miR-21 is also involved. Thus, the molecular mechanisms elucidated in this work might provide the basic information for stem cell therapy for vascular disease, e.g. tissue engineering and endothelial repair in damaged vessels.

Yikuan Chen, Mei Mei Wong, Paola Campagnolo, Russell Simpson, Bernhard Winkler, Andriani Margariti, Yanhua Hu, Qingbo Xu (2013)Adventitial Stem Cells in Vein Grafts Display Multilineage Potential That Contributes to Neointimal Formation, In: Arteriosclerosis, thrombosis, and vascular biology33(8)pp. 1844-1851 Lippincott Williams & Wilkins

Objective This study was designed to carry out the characterization of stem cells within the adventitia and to elucidate their functional role in the pathogenesis of vein graft atherosclerosis. Approach and Results A mouse vein graft model was used to investigate the functional role of adventitial stem/progenitor cells on atherosclerosis. The adventitia of vein grafts underwent significant remodeling during early stages of vessel grafting and displayed markedly heterogeneous cell compositions. Immunofluorescence staining indicated a significant number of stem cell antigen-1-positive cells that were closely located to vasa vasorum. In vitro clonogenic assays demonstrated 1% to 11% of growing rates from adventitial cell cultures, most of which could be differentiated into smooth muscle cells (SMCs). These stem cell antigen-1-positive cells also displayed a potential to differentiate into adipogenic, osteogenic, or chondrogenic lineages in vitro. In light of the proatherogenic roles of SMCs in atherosclerosis, we focused on the functional roles of progenitor-SMC differentiation, in which we subsequently demonstrated that it was driven by direct interaction of the integrin/collagen IV axis. The ex vivo bioreactor system revealed the migratory capacity of stem cell antigen-1-positive progenitor cells into the vessel wall in response to stromal cell-derived factor-1. Stem cell antigen-1-positive cells that were applied to the outer layer of vein grafts showed enhanced atherosclerosis in apolipoprotein E-deficient mice, which contributed to approximate to 30% of neointimal SMCs. Conclusions We demonstrate that during pathological conditions in vein grafting, the adventitia harbors stem/progenitor cells that can actively participate in the pathogenesis of vascular disease via differentiation into SMCs.

Mei Mei Wong, Yikuan Chen, Andriani Margariti, Bernhard Winkler, Paola Campagnolo, Claire Potter, Yanhua Hu, Qingbo Xu (2014)Macrophages Control Vascular Stem/Progenitor Cell Plasticity Through Tumor Necrosis Factor-alpha-Mediated Nuclear Factor-kappa B Activation, In: Arteriosclerosis, thrombosis, and vascular biology34(3)pp. 635-643 Lippincott Williams & Wilkins

Objective Vascular lineage differentiation of stem/progenitor cells can contribute to both tissue repair and exacerbation of vascular diseases such as in vein grafts. The role of macrophages in controlling vascular progenitor differentiation is largely unknown and may play an important role in graft development. This study aims to identify the role of macrophages in vascular stem/progenitor cell differentiation and thereafter elucidate the mechanisms that are involved in the macrophage- mediated process. Approach and Results We provide in vitro evidence that macrophages can induce endothelial cell (EC) differentiation of the stem/progenitor cells while simultaneously inhibiting their smooth muscle cell differentiation. Mechanistically, both effects were mediated by macrophage-derived tumor necrosis factor- (TNF-) via TNF- receptor 1 and canonical nuclear factor-B activation. Although the overexpression of p65 enhanced EC (or attenuated smooth muscle cell) differentiation, p65 or TNF- receptor 1 knockdown using lentiviral short hairpin RNA inhibited EC (or rescued smooth muscle cell) differentiation in response to TNF-. Furthermore, TNF--mediated EC differentiation was driven by direct binding of nuclear factor-B (p65) to specific VE-cadherin promoter sequences. Subsequent experiments using an ex vivo decellularized vessel scaffold confirmed an increase in the number of ECs and reduction in smooth muscle cell marker expression in the presence of TNF-. The lack of TNF- in a knockout mouse model of vein graft decreased endothelialization and significantly increased thrombosis formation. Conclusions Our study highlights the role of macrophages in directing vascular stem/progenitor cell lineage commitment through TNF--mediated TNF- receptor 1 and nuclear factor-B activation that is likely required for endothelial repair in vascular diseases such as vein graft.

Paola Campagnolo, Rajesh Katare, Paolo Madeddu (2018)Realities and misconceptions on the pericytes role in tissue repair, In: Regenerative medicine13(2)pp. 121-124 Future Medicine Ltd
Tsung-Neng Tsai, John Paul Kirton, Paola Campagnolo, Li Zhang, Qingzhong Xiao, Zhongyi Zhang, Wen Wang, Yanhua Hu, Qingbo Xu (2012)Contribution of Stem Cells to Neointimal Formation of Decellularized Vessel Grafts in a Novel Mouse Model, In: The American journal of pathology181(1)pp. 362-373 Elsevier

Artificial vessel grafts are often used for the treatment of occluded blood vessels, but neointimal lesions commonly occur. To both elucidate and quantify which cell types contribute to the developing neointima, we established a novel mouse model of restenosis by grafting a decellularized vessel to the carotid artery. Typically, the graft developed neointimal lesions after 2 weeks, resulting in lumen closure within 4 weeks. Immunohistochemical staining revealed the presence of endothelial and smooth muscle cells, monocytes, and stem/progenitor cells at 2 weeks after implantation. Explanted cultures of neointimal tissues displayed heterogeneous outgrowth in stem cell medium. These lesional cells expressed a panel of stem/progenitor markers, including c-kit, stem cell antigen-1 (Sca-1), and CD34. Furthermore, these cells showed clonogenic and multilineage differentiation capacities. Isolated Sca-1(+) cells were able to differentiate into endothelial and smooth muscle cells in response to vascular endothelial growth factor (VEGF) or platelet-derived growth factor (PDGF)-BB stimulation in vitro. In vivo, local application of VEGF to the adventitial side of the decellularized vessel increased re-endothelialization and reduced neointimal formation in samples at 4 weeks after implantation. A population of stem/progenitor cells exists within developing neointima, which displays the ability to 362 differentiate into both endothelial and smooth muscle cells and can contribute to restenosis. Our findings also indicate that drugs or cytokines that direct cell differentiation toward an endothelial lineage may be effective tools in the prevention or delay of restenosis. (Am J Pathol 2012, 181:362-373; http://dx.doi.org/10.1016/j.ajpath.2012.03.021)

Paola Campagnolo, Daniela Cesselli, Ayman Al Haj Zen, Antonio Paolo Beltrami, Nicolle Kraenkel, Rajesh Katare, Gianni Angelini, Costanza Emanueli, Paolo Madeddu (2010)Human Adult Vena Saphena Contains Perivascular Progenitor Cells Endowed With Clonogenic and Proangiogenic Potential, In: Circulation (New York, N.Y.)121(15)pp. 1735-U112 Lippincott Williams & Wilkins

Background-Clinical trials in ischemic patients showed the safety and benefit of autologous bone marrow progenitor cell transplantation. Non-bone marrow progenitor cells with proangiogenic capacities have been described, yet they remain clinically unexploited owing to their scarcity, difficulty of access, and low ex vivo expansibility. We investigated the presence, antigenic profile, expansion capacity, and proangiogenic potential of progenitor cells from the saphenous vein of patients undergoing coronary artery bypass surgery. Methods and Results-CD34-positive cells, negative for the endothelial marker von Willebrand factor, were localized around adventitial vasa vasorum. After dissection of the vein from surrounding tissues and enzymatic digestion, CD34-positive/CD31-negative cells were isolated by selective culture, immunomagnetic beads, or fluorescence-assisted cell sorting. In the presence of serum, CD34-positive/CD31-negative cells gave rise to a highly proliferative population that expressed pericyte/mesenchymal antigens together with the stem cell marker Sox2 and showed clonogenic and multilineage differentiation capacities. We called this population "saphenous vein-derived progenitor cells" (SVPs). In culture, SVPs integrated into networks formed by endothelial cells and supported angiogenesis through paracrine mechanisms. Reciprocally, endothelial cell-released factors facilitated SVP migration. These interactive responses were inhibited by Tie-2 or platelet-derived growth factor-BB blockade. Intramuscular injection of SVPs in ischemic limbs of immunodeficient mice improved neovascularization and blood flow recovery. At 14 days after transplantation, proliferating SVPs were still detectable in the recipient muscles, where they established N-cadherin-mediated physical contact with the capillary endothelium. Conclusions-SVPs generated from human vein CD34-positive/CD31-negative progenitor cells might represent a new therapeutic tool for angiogenic therapy in ischemic patients. (Circulation. 2010;121:1735-1745.)

Ciro Chiappini, Paola Campagnolo, Carina S Almeida, Nima Abbassi-Ghadi, Lesley W Chow, George B Hanna, Molly M Stevens (2015)Mapping Local Cytosolic Enzymatic Activity in Human Esophageal Mucosa with Porous Silicon Nanoneedles, In: Advanced materials (Weinheim)27(35)pp. 5147-5152

Porous silicon nanoneedles can map Cathepsin B activity across normal and tumor human esophageal mucosa. Assembling a peptide-based Cathepsin B cleavable sensor over a large array of nano-needles allows the discrimination of cancer cells from healthy ones in mixed culture. The same sensor applied to tissue can map Cathepsin B activity with high resolution across the tumor margin area of esophageal adenocarcinoma.

Paola Campagnolo, Mei Mei Wong, Qingbo Xu (2011)Progenitor cells in arteriosclerosis: good or bad guys?, In: Antioxidants & redox signaling15(4)pp. 1013-1027

Accumulating evidence indicates that the mobilization and recruitment of circulating or tissue-resident progenitor cells that give rise to endothelial cells (ECs) and smooth muscle cells (SMCs) can participate in atherosclerosis, neointima hyperplasia after arterial injury, and transplant arteriosclerosis. It is believed that endothelial progenitor cells do exist and can repair and rejuvenate the arteries under physiologic conditions; however, they may also contribute to lesion formation by influencing plaque stability in advanced atherosclerotic plaque under specific pathologic conditions. At the same time, smooth muscle progenitors, despite their capacity to expedite lesion formation during restenosis, may serve to promote atherosclerotic plaque stabilization by producing extracellular matrix proteins. This profound evidence provides support to the hypothesis that both endothelial and smooth muscle progenitors may act as a double-edged sword in the pathogenesis of arteriosclerosis. Therefore, the understanding of the regulatory networks that control endothelial and smooth muscle progenitor differentiation is undoubtedly fundamental both for basic research and for improving current therapeutic avenues for atherosclerosis. We update the progress in progenitor cell study related to the development of arteriosclerosis, focusing specifically on the role of progenitor cells in lesion formation and discuss the controversial issues that regard the origins, frequency, and impact of the progenitors in the disease.

Paola Campagnolo, Tsung-Neng Tsai, Xuechong Hong, John Paul Kirton, Po-Wah So, Andriana Margariti, Elisabetta Di Bernardini, Mei Mei Wong, Yanhua Hu, Molly M. Stevens, Qingbo Xu (2015)c-Kit plus progenitors generate vascular cells for tissue-engineered grafts through modulation of the Wnt/Klf4 pathway, In: Biomaterials60pp. 53-61 Elsevier

The development of decellularised scaffolds for small diameter vascular grafts is hampered by their limited patency, due to the lack of luminal cell coverage by endothelial cells (EC) and to the low tone of the vessel due to absence of a contractile smooth muscle cells (SMC). In this study, we identify a population of vascular progenitor c-Kit+/Sca-1- cells available in large numbers and derived from immuno-privileged embryonic stem cells (ESCs). We also define an efficient and controlled differentiation protocol yielding fully to differentiated ECs and SMCs in sufficient numbers to allow the repopulation of a tissue engineered vascular graft. When seeded ex vivo on a decellularised vessel, c-Kit+/Sca-1-derived cells recapitulated the native vessel structure and upon in vivo implantation in the mouse, markedly reduced neointima formation and mortality, restoring functional vascularisation. We showed that Kruppel-like transcription factor 4 (Klf4) regulates the choice of differentiation pathway of these cells through beta-catenin activation and was itself regulated by the canonical Wnt pathway activator lithium chloride. Our data show that ESC-derived c-Kit+/Sca-1-cells can be differentiated through a Klf4/beta-catenin dependent pathway and are a suitable source of vascular progenitors for the creation of superior tissue-engineered vessels from decellularised scaffolds. (C) 2015 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

P. Dominek, P. Campagnolo, M. H-Zadeh, N. Kraenkel, M. Chilosi, J. A. Sharman, A. Caporali, G. Mangialardi, G. Spinetti, C. Emanueli, M. Pignatelli, P. Madeddu (2010)Role of human tissue kallikrein in gastrointestinal stromal tumour invasion, In: British journal of cancer103(9)pp. 1422-1431 Springer Nature

BACKGROUND: Human tissue kallikrein (hK1) generates vasodilator kinins from kininogen and promotes angiogenesis by kinin-dependent and kinin-independent mechanisms. Here, we investigate the expression and functional relevance of hK1 in human gastrointestinal stromal tumour (GIST). METHODS: Vascularisation and hK1 expression of GIST samples were assessed by immunohistochemistry. In two GIST cell lines, hK1 expression was assessed by PCR, and hK1 protein levels and activity were measured by ELISA and an amidolytic assay, respectively. The effect of hK1 silencing, inhibition or overexpression on GIST cell proliferation, migration and paracrine induction of angiogenesis was studied. Finally, local and systemic levels of hK1 were assessed in mice injected with GIST cells. RESULTS: Human tissue kallikrein was detected in 19 out of 22 human GIST samples. Moreover, GIST cells express and secrete active hK1. Titration of hK1 demonstrated its involvement in GIST invasive behaviour, but not proliferation. Furthermore, hK1 released by GIST cells promoted endothelial cell migration and network formation through kinin-dependent mechanisms. Gastrointestinal stromal tumour implantation in nude mice resulted in local and systemic hK1 expression proportional to tumour dimension. CONCLUSIONS: Human tissue kallikrein is produced and released by GIST and participates in tumour invasion. Further studies are needed to validate hK1 as a diagnostic biomarker and therapeutic target in GIST. British Journal of Cancer (2010) 103, 1422-1431. doi: 10.1038/sj.bjc.6605906 www.bjcancer.com Published online 21 September 2010 (C) 2010 Cancer Research UK

Paola Campagnolo, Adam J. Gormley, Lesley W. Chow, Anne Géraldine Guex, Paresh A. Parmar, Jennifer L. Puetzer, Joseph A.M. Steele, Paolo Madeddu, Molly M. Stevens (2016)Research Data Supporting "Pericyte Seeded Dual Peptide Scaffold With Improved Endothelialization For Vascular Graft Tissue Engineering" Zenodo

Raw research data supporting the paper: Campagnolo, P. et al., Pericyte seeded dual peptide scaffold with improved endothelialization for vascular graft tissue engineering, 2016, Advanced Healthcare Materials, 5(23), 3046-3055.

Daniella Lefteri, Steven Bryden, Marieke Pingen, Sandra Terry, Emily Beswick, Georgi Georgiev, Marleen Van Der Laan, Valeria Mastrullo, Paola Campagnolo, Robert Waterhouse, Margus Varjak, Andres Merits, Rennos Fragkoudis, Stephen Griffin, Kave Shams, Emilie Pondeville, Clive Mckimmie Mosquito saliva sialokinin-dependent enhancement of arbovirus infection through endothelial barrier leakage, In: bioRxiv Cold Spring Harbor Laboratory Press

Summary Viruses transmitted by Aedes mosquitoes constitute an increasingly important global health burden. Defining common determinants of host susceptibility to this large group of heterogenous pathogens are key for informing the rational design of new pan-viral medicines. Infection of the vertebrate host with these viruses is enhanced by the presence of mosquito saliva, a complex mixture of salivary gland-derived factors and microbiota. We show that enhancement of infection by saliva was dependent on vascular function and was independent of most anti-saliva immune responses, including to salivary microbiota. Instead, the Aedes gene product sialokinin mediated enhancement of virus infection through a rapid reduction in endothelial barrier integrity. Sialokinin is unique within the insect world as having vertebrate-like tachykinin sequence and is absent from non-vector competent Anopheles mosquitoes, whose saliva was not pro-viral and did not induce similar vascular permeability. Therapeutic strategies targeting sialokinin have potential to limit disease severity following infection with Aedes mosquito-borne viruses. Competing Interest Statement The authors have declared no competing interest.

Rajesh Katare, Federica Riu, Kathryn Mitchell, Miriam Gubernator, Paola Campagnolo, Yuxin Cui, Orazio Fortunato, Elisa Avolio, Daniela Cesselli, Antonio Paolo Beltrami, Gianni Angelini, Costanza Emanueli, Paolo Madeddu (2011)Transplantation of Human Pericyte Progenitor Cells Improves the Repair of Infarcted Heart Through Activation of an Angiogenic Program Involving Micro-RNA-132, In: Circulation research109(8)pp. 894-U191 Lippincott Williams & Wilkins

Rationale: Pericytes are key regulators of vascular maturation, but their value for cardiac repair remains unknown. Objective: We investigated the therapeutic activity and mechanistic targets of saphenous vein-derived pericyte progenitor cells (SVPs) in a mouse myocardial infarction (MI) model. Methods and Results: SVPs have a low immunogenic profile and are resistant to hypoxia/starvation (H/S). Transplantation of SVPs into the peri-infarct zone of immunodeficient CD1/Foxn-1(nu/nu) or immunocompetent CD1 mice attenuated left ventricular dilatation and improved ejection fraction compared to vehicle. Moreover, SVPs reduced myocardial scar, cardiomyocyte apoptosis and interstitial fibrosis, improved myocardial blood flow and neovascularization, and attenuated vascular permeability. SVPs secrete vascular endothelial growth factor A, angiopoietin-1, and chemokines and induce an endogenous angiocrine response by the host, through recruitment of vascular endothelial growth factor B expressing monocytes. The association of donor-and recipient-derived stimuli activates the proangiogenic and prosurvival Akt/eNOS/Bcl-2 signaling pathway. Moreover, microRNA-132 (miR-132) was constitutively expressed and secreted by SVPs and remarkably upregulated, together with its transcriptional activator cyclic AMP response element-binding protein, on stimulation by H/S or vascular endothelial growth factor B. We next investigated if SVP-secreted miR-132 acts as a paracrine activator of cardiac healing. In vitro studies showed that SVP conditioned medium stimulates endothelial tube formation and reduces myofibroblast differentiation, through inhibition of Ras-GTPase activating protein and methyl-CpG-binding protein 2, which are validated miR-132 targets. Furthermore, miR-132 inhibition by antimiR-132 decreased SVP capacity to improve contractility, reparative angiogenesis, and interstitial fibrosis in infarcted hearts. Conclusion: SVP transplantation produces long-term improvement of cardiac function through a novel paracrine mechanism involving the secretion of miR-132 and inhibition of its target genes. (Circ Res. 2011;109:894-906.)

Libero Vitiello, Nicola Bassi, Paola Campagnolo, Eva Zaccariotto, Gianluca Occhi, Alberto Malerba, Sarah Pigozzo, Carlo Reggiani, Simonetta Ausoni, Tania Zaglia, Piergiorgio Gamba, Maurizio D. Baroni, Andrea P. Ditadi (2008)In vivo delivery of naked antisense oligos in aged mdx mice: Analysis of dystrophin restoration in skeletal and cardiac muscle, In: Neuromuscular disorders : NMD18(8)pp. 597-605 Elsevier B.V

Antisense-mediated exon skipping holds great potential for the treatment of DMD. In mdx mice, functional recovery of skeletal muscle has been obtained upon systemic delivery of “naked” oligonucleotides or viral vectors encoding for antisense snRNAs. However, amongst the studies reported so far, which used either neonatal or young adult animals – only one achieved dystrophin restoration in cardiac muscle, using an adeno-associated vector. Here we report the in vivo delivery of morpholino oligos in aged mdx mice, both in skeletal muscle, via intra-arterial injection, and in cardiac muscle, via intra-muscular injection. Localized intra-arterial delivery yielded high levels of dystrophin restoration and just two doses of 100 μg each resulted into detectable force recovery in the EDL muscles of treated limbs. On the other hand, upon intra-cardiac injections in the left ventricle wall the skipping effect was much lower than what obtained in tibialis anterior muscles injected with comparable amounts of oligos. This latter finding suggests that even upon direct delivery antisense-mediated dystrophin restoration in cardiac muscle might suffer from limitations that do not exist in skeletal muscle.

Oliver A. Stone, Christine Richer, Costanza Emanueli, Vincent van Weel, Paul H. A. Quax, Rajesh Katare, Nicolle Kraenkel, Paola Campagnolo, Luciola S. Barcelos, Mauro Siragusa, Graciela B. Sala-Newby, Danila Baldessari, Marina Mione, Marie P. Vincent, Andrew V. Benest, Ayman Al Haj Zen, Julien Gonzalez, David O. Bates, Francois Alhenc-Gelas, Paolo Madeddu (2009)Critical Role of Tissue Kallikrein in Vessel Formation and Maturation Implications for Therapeutic Revascularization, In: Arteriosclerosis, thrombosis, and vascular biology29(5)pp. 657-U91 Lippincott Williams & Wilkins

Objective-Human Tissue Kallikrein (hKLK1) overexpression promotes an enduring neovascularization of ischemic tissue, yet the cellular mechanisms of hKLK1-induced arteriogenesis remain unknown. Furthermore, no previous study has compared the angiogenic potency of hKLK1, with its loss of function polymorphic variant, rs5515 (R53H), which possesses reduced kinin-forming activity. Methods and Results-Here, we demonstrate that tissue kallikrein knockout mice (KLK1(-/-)) show impaired muscle neovascularization in response to hindlimb ischemia. Gene-transfer of wild-type Ad.hKLK1 but not Ad.R53H-hKLK1 was able to rescue this defect. Similarly, in the rat mesenteric assay, Ad.hKLK1 induced a mature neovasculature with increased vessel diameter through kinin-B-2 receptor-mediated recruitment of pericytes and vascular smooth muscle cells, whereas Ad.R53H-hKLK1 was ineffective. Moreover, hKLK1 but not R53H-hKLK1 overexpression in the zebrafish induced endothelial precursor cell migration and vascular remodeling. Furthermore, Ad. hKLK1 activates metalloproteinase (MMP) activity in normoperfused muscle and fails to promote reparative neovascularization in ischemic MMP9(-/-) mice, whereas its proarteriogenic action was preserved in ApoE(-/-) mice, an atherosclerotic model of impaired angiogenesis. Conclusions-These results demonstrate the fundamental role of endogenous Tissue Kallikrein in vascular repair and provide novel information on the cellular and molecular mechanisms responsible for the robust arterialization induced by hKLK1 overexpression. (Arterioscler Thromb Vasc Biol. 2009; 29: 657-664.)

Paolo Madeddu, Nicolle Kraenkel, Luciola S. Barcelos, Mauro Siragusa, Paola Campagnolo, Atsuhiko Oikawa, Andrea Caporali, Andrew Herman, Ornella Azzolino, Laura Barberis, Alessia Perino, Federico Damilano, Costanza Emanueli, Emilio Hirsch (2008)Phosphoinositide 3-kinase gamma gene knockout impairs postischemic neovascularization and endothelial progenitor cell functions, In: Arteriosclerosis, thrombosis, and vascular biology28(1)pp. 68-76 Lippincott Williams & Wilkins

Objective-We evaluated whether phosphatidylinositol 3-kinase gamma (PI3K gamma) plays a role in reparative neovascularization and endothelial progenitor cell (EPC) function. Methods and Results-Unilateral limb ischemia was induced in mice lacking the PI3K gamma gene (PI3K gamma(-/-)) or expressing a catalytically inactive mutant (PI3K gamma(KD/KD)) and wild-type controls (WT). Capillarization and arteriogenesis were reduced in PI3K gamma(-/-) ischemic muscles resulting in delayed reperfusion compared with WT, whereas reparative neovascularization was preserved in PI3K gamma(KD/KD). In PI3K gamma(-/-) muscles, endothelial cell proliferation was reduced, apoptosis was increased, and interstitial space was infiltrated with leukocytes but lacked cKit(+) progenitor cells that in WT muscles typically surrounded arterioles. PI3K gamma is constitutively expressed by WT EPCs, with expression levels being upregulated by hypoxia. PI3K gamma(-/-) EPCs showed a defect in proliferation, survival, integration into endothelial networks, and migration toward SDF-1. The dysfunctional phenotype was associated with nuclear constraining of FOXO1, reduced Akt and eNOS phosphorylation, and decreased nitric oxide ( NO) production. Pretreatment with an NO donor corrected the migratory defect of PI3K gamma(-/-) EPCs. PI3K gamma(KD/KD) EPCs showed reduced Akt phosphorylation, but constitutive activation of eNOS and preserved proliferation, survival, and migration. Conclusions-We newly demonstrated that PI3K gamma modulates angiogenesis, arteriogenesis, and vasculogenesis by mechanisms independent from its kinase activity.

Livia Santos, Gregor Fuhrmann, Maya Juenet, Nadav Amdursky, Christine-Maria Horejs, Paola Campagnolo, Molly M. Stevens (2015)Extracellular Stiffness Modulates the Expression of Functional Proteins and Growth Factors in Endothelial Cells, In: Advanced healthcare materials4(14)pp. 2056-2063 Wiley

Angiogenesis, the formation of blood vessels from pre-existing ones, is of vital importance during the early stages of bone healing. Extracellular stiffness plays an important role in regulating endothelial cell behavior and angiogenesis, but how this mechanical cue affects proliferation kinetics, gene regulation, and the expression of proteins implicated in angiogenesis and bone regeneration remains unclear. Using collagen-coated polyacrylamide (PAAm) hydrogels, human umbilical vein endothelial cells (HUVECs) are exposed to an environment that mimics the elastic properties of collagenous bone, and cellular proliferation and gene and protein expressions are assessed. The proliferation and gene expression of HUVECs are not differentially affected by culture on 3 or 30 kPa PAAm hydrogels, henceforth referred to as low and high stiffness gels, respectively. Although the proliferation and gene transcript levels remain unchanged, significant differences are found in the expressions of functional proteins and growth factors implicated both in the angiogenic and osteogenic processes. The down-regulation of the vascular endothelial growth factor receptor-2 protein with concomitant over-expression of caveolin-1, wingless-type 2, bone morphogenic protein 2, and basic fibroblast growth factor on the high stiffness PAAm hydrogel suggests that rigidity has a pro-angiogenic effect with inherent benefits for bone regeneration.

Paola Campagnolo, Xuechong Hong, Elisabetta Di Bernardini, Ioannis Smyrnias, Yanhua Hu, Qingbo Xu (2015)Resveratrol-Induced Vascular Progenitor Differentiation towards Endothelial Lineage via MiR-21/Akt/beta-Catenin Is Protective in Vessel Graft Models, In: PloS one10(5)pp. e0125122-e0125122 Public Library Science

Background and Purpose Vessel graft failure is typically associated with arteriosclerosis, in which endothelial dysfunction/ damage is a key event. Resveratrol has been shown to possess cardioprotective capacity and to reduce atherosclerosis. We aimed to study the influence of resveratrol on the behavior of resident stem cells that may contribute to graft arteriosclerosis. Experimental Approach Vascular resident progenitor cells and embryonic stem cells were treated with resveratrol under differentiating conditions and endothelial markers expression was evaluated. Expression of miR-21 and beta-catenin was also tested and exogenously modified. Effects of resveratrol treatment in an ex vivo re-endothelialization model and on mice undergone vascular graft were evaluated. Key Results Resveratrol induced expression of endothelial markers such as CD31, VE-cadherin and eNOS in both progenitor and stem cells. We demonstrated that resveratrol significantly reduced miR-21 expression, which in turn reduced Akt phosphorylation. This signal cascade diminished the amount of nuclear beta-catenin, inducing endothelial marker expression and increasing tube-like formation by progenitor cells. Both the inhibition of miR-21 and the knockdown of beta-catenin were able to recapitulate the effect of resveratrol application. Ex vivo, progenitor cells treated with resveratrol produced better endothelialization of the decellularized vessel. Finally, in a mouse model of vessel graft, a resveratrol-enhanced diet was able to reduce lesion formation. Conclusions and Implications We provide the first evidence that oral administration of resveratrol can reduce neointimal formation in a model of vascular graft and elucidated the underpinning miR-21/Akt/beta-catenin dependent mechanism. These findings may support the beneficial effect of resveratrol supplementation for graft failure prevention.

Rolando Matos, Davide Maselli, John Mcvey, Christian Heiss, Paola Campagnolo 3D printed bioreactor enabling the physiological culture and the study of pathological processes in large vessels, In: bioRxiv Cold Spring Harbor Laboratory Press

Routine cardiovascular interventions such as balloon angioplasty, result in vascular activation and remodeling, often requiring re-interventions. 2D in vitro culture models and small animal experiments have enabled the discovery of important molecular and cellular pathways involved in this process, however the clinical translation of these results is often underwhelming. There is a critical need for an ex vivo model representative of the human vascular physiology and encompassing the complexity of the vascular wall and the physical forces regulating its function. Vascular bioreactors for ex vivo culture of large vessels are viable alternatives, but their custom-made design and insufficient characterization often hinders the reproducibility of the experiments. The objective of the study was to design and validate a novel 3D printed cost-efficient and versatile perfusion system, capable of sustaining the viability and functionality of large porcine arteries for 7 days and enabling monitoring of post-injury remodeling. MultiJet Fusion 3D printing technology was used to engineer the EasyFlow insert, converting a conventional 50 ml centrifuge tube into a mini bioreactor. Porcine carotid arteries either left untreated or injured with a conventional angioplasty balloon, were cultured under pulsatile flow for up to 7 days. Pressure, heart rate, medium viscosity and shear conditions were adjusted to represent the typical arterial physiology. Tissue viability, cell activation and matrix remodeling were analyzed by immunohistochemistry, and vascular function was monitored by duplex ultrasound. Physiological blood flow conditions in the EasyFlow bioreactor preserved endothelial coverage and smooth muscle organization and extracellular matrix structure in the vessel wall, as compared to static culture. Injured arteries presented hallmarks of early remodeling, such as intimal denudation, smooth muscle cell disarray and media/adventitia activation. Duplex ultrasound confirmed physiological hemodynamic conditions, dose-dependent vasodilator response to nitroglycerin in untreated vessels and impaired dilator response in angioplastied vessels. We here validate a low-cost, robust and reproducible system to study large blood vessels physiopathology, laying the basis for future investigations into the pathological remodeling of blood vessels and creating a platform to test novel therapies and devices ex vivo in a patient relevant system. Competing Interest Statement The authors have declared no competing interest.

Rolando Szilveszter Matos, Akram Jassim Jawad Jawad, Davide Maselli, John H. McVey, Christian Heiss, Paola Campagnolo (2023)Ex Vivo Perfusion Culture of Large Blood Vessels in a 3D Printed Bioreactor, In: JoVE MyJove Corporation

Vascular disease forms the basis of most cardiovascular diseases (CVDs), which remain the primary cause of mortality and morbidity worldwide. Efficacious surgical and pharmacological interventions to prevent and treat vascular disease are urgently needed. In part, the shortage of translational models limits the understanding of the cellular and molecular processes involved in vascular disease. Ex vivo perfusion culture bioreactors provide an ideal platform for the study of large animal vessels (including humans) in a controlled dynamic environment, combining the ease of in vitro culture and the complexity of the live tissue. Most bioreactors are, however, custom manufactured and therefore difficult to adopt, limiting the reproducibility of the results. This paper presents a 3D printed system that can be easily produced and applied in any biological lab, and provides a detailed protocol for its setup, enabling users operation. This innovative and reproducible ex vivo perfusion culture system enables the culture of blood vessels for up to 7 days in physiological conditions. We expect that adopting a standardized perfusion bioreactor will support a better understanding of physiological and pathological processes in large animal blood vessels and accelerate the discovery of new therapeutics.

GAVRIELLE REBECCA UNTRACHT, ROLANDO SZILVESZTER MATOS, NIKOLAOS DIKAIOS, MARIAM BAPIR, ABDULLAH KHAN DURRANI, Teemapron Butsabong, PAOLA CAMPAGNOLO, DAVID SAMPSON, CHRISTIAN HEISS, DANUTA SAMPSON (2021)OCTAVA: An open-source toolbox for quantitative analysis of optical coherence tomography angiography images, In: Plos One16(12)pp. 1-22 Public Library of Science

Optical coherence tomography angiography (OCTA) performs non-invasive visualization and characterization of microvasculature in research and clinical applications mainly in ophthalmology and dermatology. A wide variety of instruments, imaging protocols, processing methods and metrics have been used to describe the microvasculature, such that comparing different study outcomes is currently not feasible. With the goal of contributing to standardization of OCTA data analysis, we report a user-friendly, open-source toolbox, OCTAVA (OCTA Vascular Analyzer), to automate the pre-processing, segmentation, and quantitative analysis of en face OCTA maximum intensity projection images in a standardized workflow. We present each analysis step, including optimization of filtering and choice of segmentation algorithm, and definition of metrics. We perform quantitative analysis of OCTA images from different commercial and non-commercial instruments and samples and show OCTAVA can accurately and reproducibly determine metrics for characterization of microvasculature. Wide adoption could enable studies and aggregation of data on a scale sufficient to develop reliable microvascular biomarkers for early detection, and to guide treatment, of microvascular disease.