Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Biofabrication ; 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38986455

ABSTRACT

Over the past three decades, cell therapy development has fallen short of expectations, with many cellular sources demonstrating a "Janus effect" and raising safety concerns. Extracellular vesicles (EVs), supported by advanced technologies, present a promising avenue in regenerative medicine, offering benefits such as immune tolerance and avoidance of negative aspects associated with cell transplants. Our previous research showcased enhanced and organized subcutaneous vascularization using three-dimensional bioprinted patches containing HUVEC-derived EVs in immunodeficient animal models. In this context, stress conditions on the cells of origin further boosted the EVs' neoangiogenic potential. Since neovascularization is the first regenerative target requiring restoration, the present study aims to complement our previous work by employing an injectable gelatin methacrylate (GelMA) hydrogel functionalized with HUVEC-derived EVs in a pathological condition of acute myocardial infarction. This bioactive hydrogel resulted in reduced fibrosis, improved contractility, and promoted angiogenesis, showing promise in countering tissue deterioration and addressing vascular deficits. Moreover, the molecular characterization of EVs through miRNome and proteomic analyses further supports their potential as bio-additives for hydrogel functionalization. This cell-free approach mitigates immune rejection and oncogenic risks, offering innovative therapeutic advantages.

2.
Cells ; 12(5)2023 02 22.
Article in English | MEDLINE | ID: mdl-36899835

ABSTRACT

AL amyloidosis is caused by the misfolding of immunoglobulin light chains leading to an impaired function of tissues and organs in which they accumulate. Due to the paucity of -omics profiles from undissected samples, few studies have addressed amyloid-related damage system wide. To fill this gap, we evaluated proteome changes in the abdominal subcutaneous adipose tissue of patients affected by the AL isotypes κ and λ. Through our retrospective analysis based on graph theory, we have herein deduced new insights representing a step forward from the pioneering proteomic investigations previously published by our group. ECM/cytoskeleton, oxidative stress and proteostasis were confirmed as leading processes. In this scenario, some proteins, including glutathione peroxidase 1 (GPX1), tubulins and the TRiC complex, were classified as biologically and topologically relevant. These and other results overlap with those already reported for other amyloidoses, supporting the hypothesis that amyloidogenic proteins could induce similar mechanisms independently of the main fibril precursor and of the target tissues/organs. Of course, further studies based on larger patient cohorts and different tissues/organs will be essential, which would be a key point that would allow for a more robust selection of the main molecular players and a more accurate correlation with clinical aspects.


Subject(s)
Immunoglobulin Light-chain Amyloidosis , Humans , Proteomics/methods , Retrospective Studies , Biopsy , Subcutaneous Fat/metabolism
3.
Int J Mol Sci ; 23(17)2022 Aug 23.
Article in English | MEDLINE | ID: mdl-36076927

ABSTRACT

Before entering human clinical studies to evaluate their safety and effectiveness, new drugs and novel medical treatments are subject to extensive animal testing that are expensive and time-consuming. By contrast, advanced technologies enable the development of animal-free models that allow the efficacy of innovative therapies to be studied without sacrificing animals, while providing helpful information and details. We report on the powerful combination of 3D bioprinting (3DB) and photo-thermal therapy (PTT) applications. To this end, we realize a 3DB construct consisting of glioblastoma U87-MG cells in a 3D geometry, incorporating biomimetic keratin-coated gold nanoparticles (Ker-AuNPs) as a photo-thermal agent. The resulting plasmonic 3DB structures exhibit a homogeneous cell distribution throughout the entire volume while promoting the localization of Ker-AuNPs within the cells. A 3D immunofluorescence assay and transmission electron microscopy (TEM) confirm the uniform distribution of fluorescent-labeled Ker-AuNPs in the volume and their capability to enter the cells. Laser-assisted (λ = 532 nm) PTT experiments demonstrate the extraordinary ability of Ker-AuNPs to generate heating, producing the highest temperature rise of about 16 °C in less than 2 min.


Subject(s)
Glioblastoma , Hyperthermia, Induced , Metal Nanoparticles , Photothermal Therapy , Biomimetic Materials , Glioblastoma/therapy , Gold/chemistry , Humans , Keratins/chemistry , Metal Nanoparticles/chemistry , Photothermal Therapy/methods
4.
Cell Prolif ; 55(11): e13312, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35946052

ABSTRACT

OBJECTIVES: Extracellular vesicles (EVs) are key biological mediators of several physiological functions within the cell microenvironment. Platelets are the most abundant source of EVs in the blood. Similarly, platelet lysate (PL), the best platelet derivative and angiogenic performer for regenerative purposes, is enriched of EVs, but their role is still too poorly discovered to be suitably exploited. Here, we explored the contribution of the EVs in PL, by investigating the angiogenic features extrapolated from that possessed by PL. METHODS: We tested angiogenic ability and molecular cargo in 3D bioprinted models and by RNA sequencing analysis of PL-derived EVs. RESULTS: A subset of small vesicles is highly represented in PL. The EVs do not retain aggregation ability, preserving a low redox state in human umbilical vein endothelial cells (HUVECs) and increasing the angiogenic tubularly-like structures in 3D endothelial bioprinted constructs. EVs resembled the miRNome profile of PL, mainly enriched with small RNAs and a high amount of miR-126, the most abundant angiogenic miRNA in platelets. The transfer of miR-126 by EVs in HUVEC after the in vitro inhibition of the endogenous form, restored angiogenesis, without involving VEGF as a downstream target in this system. CONCLUSION: PL is a biological source of available EVs with angiogenic effects involving a miRNAs-based cargo. These properties can be exploited for targeted molecular/biological manipulation of PL, by potentially developing a product exclusively manufactured of EVs.


Subject(s)
Extracellular Vesicles , MicroRNAs , Humans , Human Umbilical Vein Endothelial Cells , MicroRNAs/genetics , Neovascularization, Pathologic , Blood Platelets
5.
Int J Mol Sci ; 23(14)2022 Jul 18.
Article in English | MEDLINE | ID: mdl-35887248

ABSTRACT

The cellular heterogeneity of the tumor environment of breast cancer (BC) is extremely complex and includes different actors such as neoplastic, stromal, and immunosuppressive cells, which contribute to the chemical and mechanical modification of the environment surrounding the tumor-exasperating immune-escaping mechanisms. In addition to molecular signals that make the tumor microenvironment (TME) unacceptable for the penetrance of the immune system, the physical properties of tumoral extracellular matrix (tECM) also have carved out a fundamental role in the processes of the protection of the tumor niche. Tumor-associated macrophages (TAMs), with an M2 immunosuppressive phenotype, are important determinants for the establishment of a tumor phenotype excluded from T cells. NF-κB transcription factors orchestrate innate immunity and represent the common thread between inflammation and cancer. Many studies have focused on canonical activation of NF-κB; however, activation of non-canonical signaling predicts poor survival and resistance to therapy. In this scenario, we demonstrated the existence of an unusual association of NF-κB components in TAMs that determines the deposition of HSPG2 that affects the stiffness of tECM. These results highlight a new mechanism counterbalanced between physical factors and a new perspective of mechano-pathology to be targeted to counteract immune evasion in BC.


Subject(s)
NF-kappa B , Neoplasms , Humans , Macrophages , Neoplasms/pathology , Tumor Microenvironment , Tumor-Associated Macrophages
6.
Cardiovasc Res ; 118(8): 1872-1884, 2022 06 29.
Article in English | MEDLINE | ID: mdl-34254111

ABSTRACT

Alterations in the DMD gene, which codes for the protein dystrophin, cause forms of dystrophinopathies such as Duchenne muscular dystrophy, an X-linked disease. Cardiomyopathy linked to DMD mutations is becoming the leading cause of death in patients with dystrophinopathy. Since phenotypic pathophysiological mechanisms are not fully understood, the improvement and development of new disease models, considering their relative advantages and disadvantages, is essential. The application of genetic engineering approaches on induced pluripotent stem cells, such as gene-editing technology, enables the development of physiologically relevant human cell models for in vitro dystrophinopathy studies. The combination of induced pluripotent stem cells-derived cardiovascular cell types and 3D bioprinting technologies hold great promise for the study of dystrophin-linked cardiomyopathy. This combined approach enables the assessment of responses to physical or chemical stimuli, and the influence of pharmaceutical approaches. The critical objective of in vitro microphysiological systems is to more accurately reproduce the microenvironment observed in vivo. Ground-breaking methodology involving the connection of multiple microphysiological systems comprised of different tissues would represent a move toward precision body-on-chip disease modelling could lead to a critical expansion in what is known about inter-organ responses to disease and novel therapies that have the potential to replace animal models. In this review, we will focus on the generation, development, and application of current cellular, animal, and potential for bio-printed models, in the study of the pathophysiological mechanisms underlying dystrophin-linked cardiomyopathy in the direction of personalized medicine.


Subject(s)
Cardiomyopathies , Induced Pluripotent Stem Cells , Muscular Dystrophy, Duchenne , Animals , Cardiomyopathies/genetics , Cardiomyopathies/therapy , Dystrophin/genetics , Dystrophin/metabolism , Heart , Induced Pluripotent Stem Cells/metabolism , Muscular Dystrophy, Duchenne/genetics
7.
Dis Model Mech ; 14(12)2021 12 01.
Article in English | MEDLINE | ID: mdl-34796900

ABSTRACT

Large-animal models for Duchenne muscular dystrophy (DMD) are crucial for the evaluation of diagnostic procedures and treatment strategies. Pigs cloned from male cells lacking DMD exon 52 (DMDΔ52) exhibit molecular, clinical and pathological hallmarks of DMD, but die before sexual maturity and cannot be propagated by breeding. Therefore, we generated female DMD+/- carriers. A single founder animal had 11 litters with 29 DMDY/-, 34 DMD+/- as well as 36 male and 29 female wild-type offspring. Breeding with F1 and F2 DMD+/- carriers resulted in an additional 114 DMDY/- piglets. With intensive neonatal management, the majority survived for 3-4 months, providing statistically relevant cohorts for experimental studies. Pathological investigations and proteome studies of skeletal muscles and myocardium confirmed the resemblance to human disease mechanisms. Importantly, DMDY/- pigs displayed progressive myocardial fibrosis and increased expression of connexin-43, associated with significantly reduced left ventricular ejection fraction, at 3 months. Furthermore, behavioral tests provided evidence for impaired cognitive ability. Our breeding cohort of DMDΔ52 pigs and standardized tissue repositories provide important resources for studying DMD disease mechanisms and for testing novel treatment strategies.


Subject(s)
Cardiomyopathies , Muscular Dystrophy, Duchenne , Animals , Cardiomyopathies/pathology , Female , Humans , Male , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/pathology , Stroke Volume , Swine , Ventricular Function, Left
8.
Int J Mol Sci ; 22(11)2021 May 30.
Article in English | MEDLINE | ID: mdl-34070750

ABSTRACT

The immune system is a fine modulator of the tumor biology supporting or inhibiting its progression, growth, invasion and conveys the pharmacological treatment effect. Tumors, on their side, have developed escaping mechanisms from the immune system action ranging from the direct secretion of biochemical signals to an indirect reaction, in which the cellular actors of the tumor microenvironment (TME) collaborate to mechanically condition the extracellular matrix (ECM) making it inhospitable to immune cells. TME is composed of several cell lines besides cancer cells, including tumor-associated macrophages, cancer-associated fibroblasts, CD4+ and CD8+ lymphocytes, and innate immunity cells. These populations interface with each other to prepare a conservative response, capable of evading the defense mechanisms implemented by the host's immune system. The presence or absence, in particular, of cytotoxic CD8+ cells in the vicinity of the main tumor mass, is able to predict, respectively, the success or failure of drug therapy. Among various mechanisms of immunescaping, in this study, we characterized the modulation of the phenotypic profile of CD4+ and CD8+ cells in resting and activated states, in response to the mechanical pressure exerted by a three-dimensional in vitro system, able to recapitulate the rheological and stiffness properties of the tumor ECM.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Extracellular Matrix/immunology , Gene Expression Regulation, Neoplastic/immunology , Tumor Escape , Tumor Microenvironment/immunology , 5'-Nucleotidase/genetics , 5'-Nucleotidase/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/pathology , Cell Culture Techniques , Elastic Modulus , Extracellular Matrix/chemistry , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Humans , Hydrogels/chemistry , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocyte Activation , Mechanotransduction, Cellular , Models, Biological , NF-kappa B/genetics , NF-kappa B/immunology , Phenotype , Primary Cell Culture , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Rheology , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/immunology , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment/genetics , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/pathology
9.
Biofabrication ; 13(3)2021 04 07.
Article in English | MEDLINE | ID: mdl-33434889

ABSTRACT

Extracellular vesicles (EVs) have become a key tool in the biotechnological landscape due to their well-documented ability to mediate intercellular communication. This feature has been explored and is under constant investigation by researchers, who have demonstrated the important role of EVs in several research fields ranging from oncology to immunology and diagnostics to regenerative medicine. Unfortunately, there are still some limitations to overcome before clinical application, including the inability to confine the EVs to strategically defined sites of interest to avoid side effects. In this study, for the first time, EV application is supported by 3D bioprinting technology to develop a new strategy for applying the angiogenic cargo of human umbilical vein endothelial cell-derived EVs in regenerative medicine. EVs, derived from human endothelial cells and grown under different stressed conditions, were collected and used as bioadditives for the formulation of advanced bioinks. Afterin vivosubcutaneous implantation, we demonstrated that the bioprinted 3D structures, loaded with EVs, supported the formation of a new functional vasculaturein situ, consisting of blood-perfused microvessels recapitulating the printed pattern. The results obtained in this study favour the development of new therapeutic approaches for critical clinical conditions, such as the need for prompt revascularization of ischaemic tissues, which represent the fundamental substrate for advanced regenerative medicine applications.


Subject(s)
Bioprinting , Extracellular Vesicles , Printing, Three-Dimensional , Cell Communication , Human Umbilical Vein Endothelial Cells , Humans , Regenerative Medicine
10.
Cells ; 9(6)2020 06 23.
Article in English | MEDLINE | ID: mdl-32585911

ABSTRACT

The recent advances, offered by cell therapy in the regenerative medicine field, offer a revolutionary potential for the development of innovative cures to restore compromised physiological functions or organs. Adult myogenic precursors, such as myoblasts or satellite cells, possess a marked regenerative capacity, but the exploitation of this potential still encounters significant challenges in clinical application, due to low rate of proliferation in vitro, as well as a reduced self-renewal capacity. In this scenario, induced pluripotent stem cells (iPSCs) can offer not only an inexhaustible source of cells for regenerative therapeutic approaches, but also a valuable alternative for in vitro modeling of patient-specific diseases. In this study we established a reliable protocol to induce the myogenic differentiation of iPSCs, generated from pericytes and fibroblasts, exploiting skeletal muscle-derived extracellular vesicles (EVs), in combination with chemically defined factors. This genetic integration-free approach generates functional skeletal myotubes maintaining the engraftment ability in vivo. Our results demonstrate evidence that EVs can act as biological "shuttles" to deliver specific bioactive molecules for a successful transgene-free differentiation offering new opportunities for disease modeling and regenerative approaches.


Subject(s)
Extracellular Vesicles/metabolism , Induced Pluripotent Stem Cells/metabolism , Muscle Development/physiology , Muscle, Skeletal/metabolism , Adult , Animals , Cell Differentiation , Healthy Volunteers , Humans , Male , Mice , Young Adult
11.
Sci Rep ; 8(1): 13532, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30201959

ABSTRACT

The myocardium behaves like a sophisticated orchestra that expresses its true potential only if each member performs the correct task harmonically. Recapitulating its complexity within engineered 3D functional constructs with tailored biological and mechanical properties, is one of the current scientific priorities in the field of regenerative medicine and tissue engineering. In this study, driven by the necessity of fabricating advanced model of cardiac tissue, we present an innovative approach consisting of heterogeneous, multi-cellular constructs composed of Human Umbilical Vein Endothelial Cells (HUVECs) and induced pluripotent cell-derived cardiomyocytes (iPSC-CMs). Cells were encapsulated within hydrogel strands containing alginate and PEG-Fibrinogen (PF) and extruded through a custom microfluidic printing head (MPH) that allows to precisely tailor their 3D spatial deposition, guaranteeing a high printing fidelity and resolution. We obtained a 3D cardiac tissue compose of iPSC-derived CMs with a high orientation index imposed by the different defined geometries and blood vessel-like shapes generated by HUVECs which, as demonstrated by in vivo grafting, better support the integration of the engineered cardiac tissue with host's vasculature.


Subject(s)
Bioprinting/methods , Bioprosthesis , Printing, Three-Dimensional , Tissue Engineering/methods , Alginates/chemistry , Animals , Bioprinting/instrumentation , Cardiac Surgical Procedures , Cardiovascular Diseases/surgery , Cell Culture Techniques/methods , Cell Differentiation , Coronary Vessels/physiology , Fibrinogen/chemistry , Fibroblasts , Human Umbilical Vein Endothelial Cells/physiology , Humans , Hydrogels/chemistry , Induced Pluripotent Stem Cells/physiology , Mice , Mice, Inbred C57BL , Microfluidics/instrumentation , Microfluidics/methods , Models, Animal , Myocardium/cytology , Myocytes, Cardiac/physiology , Primary Cell Culture , Prosthesis Implantation , Skin/cytology , Tissue Engineering/instrumentation , Tissue Scaffolds/chemistry
12.
Cell Death Dis ; 9(2): 108, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29371598

ABSTRACT

Cardiovascular diseases (CVDs) are a major burden on the healthcare system: indeed, over two million new cases are diagnosed every year worldwide. Unfortunately, important drawbacks for the treatment of these patients derive from our current inability to stop the structural alterations that lead to heart failure, the common endpoint of many CVDs. In this scenario, a better understanding of the role of epigenetics - hereditable changes of chromatin that do not alter the DNA sequence itself - is warranted. To date, hyperacetylation of histones has been reported in hypertension and myocardial infarction, but the use of inhibitors for treating CVDs remains limited. Here, we studied the effect of the histone deacetylase inhibitor Givinostat on a mouse model of acute myocardial infarction. We found that it contributes to decrease endothelial-to-mesenchymal transition and inflammation, reducing cardiac fibrosis and improving heart performance and protecting the blood vessels from apoptosis through the modulatory effect of cardiac fibroblasts on endothelial cells. Therefore, Givinostat may have potential for the treatment of CVDs.


Subject(s)
Carbamates/pharmacology , Fibroblasts/pathology , Ventricular Remodeling/drug effects , Animals , Apoptosis/drug effects , Endothelium/drug effects , Endothelium/pathology , Epithelium/drug effects , Epithelium/pathology , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/pathology , Mice, Inbred C57BL , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...