58 results on '"Perpétua Pinto-do-Ó"'
Search Results
2. P1331: DISTINCT REACTIVITY TO ENVIRONMENTAL FACTORS OF YS VERSUS HSC DERIVED PROGENITORS DICTATE HEMATOPOIESIS IN THE MOUSE FETAL LIVER
- Author
-
Francisca Soares-Da-Silva, Marcia Peixoto, Valentin Bonnet, Gustave Ronteix, Marie-Pierre Mailhe, Charles Baroud, Perpétua Pinto-Do-Ó, and Ana Cumano
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
3. Corrigendum: Consistent long-term therapeutic efficacy of human umbilical cord matrix-derived mesenchymal stromal cells after myocardial infarction despite individual differences and transient engraftment
- Author
-
Tiago L. Laundos, Francisco Vasques-Nóvoa, Rita N. Gomes, Vasco Sampaio-Pinto, Pedro Cruz, Hélder Cruz, Jorge M. Santos, Rita N. Barcia, Perpétua Pinto-do-Ó, and Diana S. Nascimento
- Subjects
mesenchymal stromal (or stem) cells ,Wharton’s jelly ,myocardial infarction ,regeneration/repair ,umbilical cord matrix derived mesenchymal stromal cells (hUCM-MSCs) ,cell therapy ,Biology (General) ,QH301-705.5 - Published
- 2023
- Full Text
- View/download PDF
4. Human-umbilical cord matrix mesenchymal cells improved left ventricular contractility independently of infarct size in swine myocardial infarction with reperfusion
- Author
-
Luís Raposo, Rui J. Cerqueira, Sara Leite, Liliana Moreira-Costa, Tiago L. Laundos, Joana O. Miranda, Pedro Mendes-Ferreira, João Almeida Coelho, Rita N. Gomes, Perpétua Pinto-do-Ó, Diana S. Nascimento, André P. Lourenço, Nuno Cardim, and Adelino Leite-Moreira
- Subjects
umbilical-cord ,mesenchymal cells ,MSC ,myocardial infarction ,reperfusion ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
BackgroundHuman umbilical cord matrix-mesenchymal stromal cells (hUCM-MSC) have demonstrated beneficial effects in experimental acute myocardial infarction (AMI). Reperfusion injury hampers myocardial recovery in a clinical setting and its management is an unmet need. We investigated the efficacy of intracoronary (IC) delivery of xenogeneic hUCM-MSC as reperfusion-adjuvant therapy in a translational model of AMI in swine.MethodsIn a placebo-controlled trial, pot-belied pigs were randomly assigned to a sham-control group (vehicle-injection; n = 8), AMI + vehicle (n = 12) or AMI + IC-injection (n = 11) of 5 × 105 hUCM-MSC/Kg, within 30 min of reperfusion. AMI was created percutaneously by balloon occlusion of the mid-LAD. Left-ventricular function was blindly evaluated at 8-weeks by invasive pressure-volume loop analysis (primary endpoint). Mechanistic readouts included histology, strength-length relationship in skinned cardiomyocytes and gene expression analysis by RNA-sequencing.ResultsAs compared to vehicle, hUCM-MSC enhanced systolic function as shown by higher ejection fraction (65 ± 6% vs. 43 ± 4%; p = 0.0048), cardiac index (4.1 ± 0.4 vs. 3.1 ± 0.2 L/min/m2; p = 0.0378), preload recruitable stroke work (75 ± 13 vs. 36 ± 4 mmHg; p = 0.0256) and end-systolic elastance (2.8 ± 0.7 vs. 2.1 ± 0.4 mmHg*m2/ml; p = 0.0663). Infarct size was non-significantly lower in cell-treated animals (13.7 ± 2.2% vs. 15.9 ± 2.7%; Δ = −2.2%; p = 0.23), as was interstitial fibrosis and cardiomyocyte hypertrophy in the remote myocardium. Sarcomere active tension improved, and genes related to extracellular matrix remodelling (including MMP9, TIMP1 and PAI1), collagen fibril organization and glycosaminoglycan biosynthesis were downregulated in animals treated with hUCM-MSC.ConclusionIntracoronary transfer of xenogeneic hUCM-MSC shortly after reperfusion improved left-ventricular systolic function, which could not be explained by the observed extent of infarct size reduction alone. Combined contributions of favourable modification of myocardial interstitial fibrosis, matrix remodelling and enhanced cardiomyocyte contractility in the remote myocardium may provide mechanistic insight for the biological effect.
- Published
- 2023
- Full Text
- View/download PDF
5. Learning the Biochemical Basis of Axonal Guidance: Using Caenorhabditis elegans as a Model
- Author
-
Andreia Teixeira-Castro, João Carlos Sousa, Cármen Vieira, Joana Pereira-Sousa, Daniela Vilasboas-Campos, Fernanda Marques, Perpétua Pinto-do-Ó, and Patrícia Maciel
- Subjects
neuronal cell biology ,axon pathfinding ,growth cone ,molecular cues ,experimental activity ,Biology (General) ,QH301-705.5 - Abstract
Aim: Experimental models are a powerful aid in visualizing molecular phenomena. This work reports how the worm Caenorhabditis elegans (C. elegans) can be effectively explored for students to learn how molecular cues dramatically condition axonal guidance and define nervous system structure and behavior at the organism level. Summary of work: A loosely oriented observational activity preceded detailed discussions on molecules implied in axonal migration. C. elegans mutants were used to introduce second-year medical students to the deleterious effects of gene malfunctioning in neuron response to extracellular biochemical cues and to establish links between molecular function, nervous system structure, and animal behavior. Students observed C. elegans cultures and associated animal behavior alterations with the lack of function of specific axon guidance molecules (the soluble cue netrin/UNC-6 or two receptors, DCC/UNC-40 and UNC-5H). Microscopical observations of these strains, in combination with pan-neuronal GFP expression, allowed optimal visualization of severely affected neurons. Once the list of mutated genes in each strain was displayed, students could also relate abnormal patterns in axon migration/ventral and dorsal nerve cord neuron formation in C. elegans with mutated molecular components homologous to those in humans. Summary of results: Students rated the importance and effectiveness of the activity very highly. Ninety-three percent found it helpful to grasp human axonal migration, and all students were surprised with the power of the model in helping to visualize the phenomenon.
- Published
- 2023
- Full Text
- View/download PDF
6. Proteomic Identification of a Gastric Tumor ECM Signature Associated With Cancer Progression
- Author
-
Ana M. Moreira, Rui M. Ferreira, Patrícia Carneiro, Joana Figueiredo, Hugo Osório, José Barbosa, John Preto, Perpétua Pinto-do-Ó, Fátima Carneiro, and Raquel Seruca
- Subjects
extracellular matrix (ECM) ,gastric cancer ,matrisome ,biomarker ,proteomics ,Biology (General) ,QH301-705.5 - Abstract
The extracellular matrix (ECM) plays an undisputable role in tissue homeostasis and its deregulation leads to altered mechanical and biochemical cues that impact cancer development and progression. Herein, we undertook a novel approach to address the role of gastric ECM in tumorigenesis, which remained largely unexplored. By combining decellularization techniques with a high-throughput quantitative proteomics approach, we have performed an extensive characterization of human gastric mucosa, uncovering its composition and distribution among tumor, normal adjacent and normal distant mucosa. Our results revealed a common ECM signature composed of 142 proteins and indicated that gastric carcinogenesis encompasses ECM remodeling through alterations in the abundance of 24 components, mainly basement membrane proteins. Indeed, we could only identify one de novo tumor-specific protein, the collagen alpha-1(X) chain (COL10A1). Functional analysis of the data demonstrated that gastric ECM remodeling favors tumor progression by activating ECM receptors and cellular processes involved in angiogenesis and cell-extrinsic metabolic regulation. By analyzing mRNA expression in an independent GC cohort available at the TGCA, we validated the expression profile of 12 differentially expressed ECM proteins. Importantly, the expression of COL1A2, LOX and LTBP2 significantly correlated with high tumor stage, with LOX and LTBP2 further impacting patient overall survival. These findings contribute for a better understanding of GC biology and highlight the role of core ECM components in gastric carcinogenesis and their clinical relevance as biomarkers of disease prognosis.
- Published
- 2022
- Full Text
- View/download PDF
7. Establishing a Link Between Endothelial Cell Metabolism and Vascular Behaviour in a Type 1 Diabetes Mouse Model
- Author
-
Carolina Silva, Vasco Sampaio-Pinto, Sara Andrade, Ilda Rodrigues, Raquel Costa, Susana Guerreiro, Eugenia Carvalho, Perpétua Pinto-do-Ó, Diana S. Nascimento, and Raquel Soares
- Subjects
Physiology ,QP1-981 ,Biochemistry ,QD415-436 - Published
- 2019
- Full Text
- View/download PDF
8. Consistent Long-Term Therapeutic Efficacy of Human Umbilical Cord Matrix-Derived Mesenchymal Stromal Cells After Myocardial Infarction Despite Individual Differences and Transient Engraftment
- Author
-
Tiago L. Laundos, Francisco Vasques-Nóvoa, Rita N. Gomes, Vasco Sampaio-Pinto, Pedro Cruz, Hélder Cruz, Jorge M. Santos, Rita N. Barcia, Perpétua Pinto-do-Ó, and Diana S. Nascimento
- Subjects
mesenchymal stromal (or stem) cells ,Wharton's jelly ,myocardial infarction ,regeneration/repair ,umbilical cord matrix derived mesenchymal stromal cells (hUCM-MSCs) ,cell therapy ,Biology (General) ,QH301-705.5 - Abstract
Human mesenchymal stem cells gather special interest as a universal and feasible add-on therapy for myocardial infarction (MI). In particular, human umbilical cord matrix-derived mesenchymal stromal cells (UCM-MSC) are advantageous since can be easily obtained and display high expansion potential. Using isolation protocols compliant with cell therapy, we previously showed UCM-MSC preserved cardiac function and attenuated remodeling 2 weeks after MI. In this study, UCM-MSC from two umbilical cords, UC-A and UC-B, were transplanted in a murine MI model to investigate consistency and durability of the therapeutic benefits. Both cellular products improved cardiac function and limited adverse cardiac remodeling 12 weeks post-ischemic injury, supporting sustained and long-term beneficial therapeutic effect. Donor associated variability was found in the modulation of cardiac remodeling and activation of the Akt-mTOR-GSK3β survival pathway. In vitro, the two cell products displayed similar ability to induce the formation of vessel-like structures and comparable transcriptome in normoxia and hypoxia, apart from UCM-MSCs proliferation and expression differences in a small subset of genes associated with MHC Class I. These findings support that UCM-MSC are strong candidates to assist the treatment of MI whilst calling for the discussion on methodologies to characterize and select best performing UCM-MSC before clinical application.
- Published
- 2021
- Full Text
- View/download PDF
9. Bearing My Heart: The Role of Extracellular Matrix on Cardiac Development, Homeostasis, and Injury Response
- Author
-
Ana Catarina Silva, Cassilda Pereira, Ana Catarina R. G. Fonseca, Perpétua Pinto-do-Ó, and Diana S. Nascimento
- Subjects
heart ,extracellular matrix ,cardiac ontogeny ,cardiovascular diseases ,decellularization ,fibrosis ,Biology (General) ,QH301-705.5 - Abstract
The extracellular matrix (ECM) is an essential component of the heart that imparts fundamental cellular processes during organ development and homeostasis. Most cardiovascular diseases involve severe remodeling of the ECM, culminating in the formation of fibrotic tissue that is deleterious to organ function. Treatment schemes effective at managing fibrosis and promoting physiological ECM repair are not yet in reach. Of note, the composition of the cardiac ECM changes significantly in a short period after birth, concurrent with the loss of the regenerative capacity of the heart. This highlights the importance of understanding ECM composition and function headed for the development of more efficient therapies. In this review, we explore the impact of ECM alterations, throughout heart ontogeny and disease, on cardiac cells and debate available approaches to deeper insights on cell–ECM interactions, toward the design of new regenerative therapies.
- Published
- 2021
- Full Text
- View/download PDF
10. Neonatal Apex Resection Triggers Cardiomyocyte Proliferation, Neovascularization and Functional Recovery Despite Local Fibrosis
- Author
-
Vasco Sampaio-Pinto, Sílvia C. Rodrigues, Tiago L. Laundos, Elsa D. Silva, Francisco Vasques-Nóvoa, Ana C. Silva, Rui J. Cerqueira, Tatiana P. Resende, Nicola Pianca, Adelino Leite-Moreira, Gabriele D'Uva, Sólveig Thorsteinsdóttir, Perpétua Pinto-do-Ó, and Diana S. Nascimento
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Summary: So far, opposing outcomes have been reported following neonatal apex resection in mice, questioning the validity of this injury model to investigate regenerative mechanisms. We performed a systematic evaluation, up to 180 days after surgery, of the pathophysiological events activated upon apex resection. In response to cardiac injury, we observed increased cardiomyocyte proliferation in remote and apex regions, neovascularization, and local fibrosis. In adulthood, resected hearts remain consistently shorter and display permanent fibrotic tissue deposition in the center of the resection plane, indicating limited apex regrowth. However, thickening of the left ventricle wall, explained by an upsurge in cardiomyocyte proliferation during the initial response to injury, compensated cardiomyocyte loss and supported normal systolic function. Thus, apex resection triggers both regenerative and reparative mechanisms, endorsing this injury model for studies aimed at promoting cardiomyocyte proliferation and/or downplaying fibrosis. : In this article, Nascimento and colleagues demonstrate that neonatal apex resection stimulates cardiomyocyte proliferation and permanent scarring in the apex. Newly formed cardiomyocytes compensate muscle loss by resection, and resected hearts recover functional competence in adulthood. These findings endorse this model for studies aiming to block cardiac fibrosis and/or favoring CM proliferation. Keywords: neonatal apex resection, cardiac regeneration, cardiac injury response, cardiomyocyte proliferation, fibrosis, cardiac fibroblasts, extracellular matrix, neovascularization, stereology
- Published
- 2018
- Full Text
- View/download PDF
11. Transient HES5 Activity Instructs Mesodermal Cells toward a Cardiac Fate
- Author
-
Ana G. Freire, Avinash Waghray, Francisca Soares-da-Silva, Tatiana P. Resende, Dung-Fang Lee, Carlos-Filipe Pereira, Diana S. Nascimento, Ihor R. Lemischka, and Perpétua Pinto-do-Ó
- Subjects
Hes5 ,notch signaling pathway ,cardiac fate specification ,nascent mesoderm ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Notch signaling plays a role in specifying a cardiac fate but the downstream effectors remain unknown. In this study we implicate the Notch downstream effector HES5 in cardiogenesis. We show transient Hes5 expression in early mesoderm of gastrulating embryos and demonstrate, by loss and gain-of-function experiments in mouse embryonic stem cells, that HES5 favors cardiac over primitive erythroid fate. Hes5 overexpression promotes upregulation of the cardiac gene Isl1, while the hematopoietic regulator Scl is downregulated. Moreover, whereas a pulse of Hes5 instructs cardiac commitment, sustained expression after lineage specification impairs progression of differentiation to contracting cardiomyocytes. These findings establish a role for HES5 in cardiogenesis and provide insights into the early cardiac molecular network.
- Published
- 2017
- Full Text
- View/download PDF
12. Restoring heart function and electrical integrity: closing the circuit
- Author
-
Luís Miguel Monteiro, Francisco Vasques-Nóvoa, Lino Ferreira, Perpétua Pinto-do-Ó, and Diana Santos Nascimento
- Subjects
Medicine - Abstract
Abstract Cardiovascular diseases are the main cause of death in the world and are often associated with the occurrence of arrhythmias due to disruption of myocardial electrical integrity. Pathologies involving dysfunction of the specialized cardiac excitatory/conductive tissue are also common and constitute an added source of morbidity and mortality since current standard therapies withstand a great number of limitations. As electrical integrity is essential for a well-functioning heart, innovative strategies have been bioengineered to improve heart conduction and/or promote myocardial repair, based on: (1) gene and/or cell delivery; or (2) conductive biomaterials as tools for cardiac tissue engineering. Herein we aim to review the state-of-art in the area, while briefly describing the biological principles underlying the heart electrical/conduction system and how this system can be disrupted in heart disease. Suggestions regarding targets for future studies are also presented.
- Published
- 2017
- Full Text
- View/download PDF
13. Mouse HSA+ immature cardiomyocytes persist in the adult heart and expand after ischemic injury.
- Author
-
Mariana Valente, Tatiana Pinho Resende, Diana Santos Nascimento, Odile Burlen-Defranoux, Francisca Soares-da-Silva, Benoit Dupont, Ana Cumano, and Perpétua Pinto-do-Ó
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The assessment of the regenerative capacity of the heart has been compromised by the lack of surface signatures to characterize cardiomyocytes (CMs). Here, combined multiparametric surface marker analysis with single-cell transcriptional profiling and in vivo transplantation identify the main mouse fetal cardiac populations and their progenitors (PRGs). We found that CMs at different stages of differentiation coexist during development. We identified a population of immature heat stable antigen (HSA)/ cluster of differentiation 24 (CD24)+ CMs that persists throughout life and that, unlike other CM subsets, actively proliferates up to 1 week of age and engrafts cardiac tissue upon transplantation. In the adult heart, a discrete population of HSA/CD24+ CMs appears as mononucleated cells that increase in frequency after infarction. Our work identified cell surface signatures that allow the prospective isolation of CMs at all developmental stages and the detection of a subset of immature CMs throughout life that, although at reduced frequencies, are poised for activation in response to ischemic stimuli. This work opens new perspectives in the understanding and treatment of heart pathologies.
- Published
- 2019
- Full Text
- View/download PDF
14. Primary Bone Marrow Mesenchymal Stromal Cells Rescue the Axonal Phenotype of Twitcher Mice
- Author
-
Catarina Oliveira Miranda, Carla Andreia Teixeira, Vera Filipe Sousa, Telma Emanuela Santos, Márcia Almeida Liz, Ana Maio Marques, Perpétua Pinto-Do-Ó, and Mónica Mendes Sousa
- Subjects
Medicine - Abstract
Krabbe's disease (KD) is a demyelinating disorder caused by the deficiency of lysosomal galactocerebrosi-dase (GALC), affecting both the central (CNS) and the peripheral nervous system (PNS). A current therapy, hematopoietic stem cell transplantation (HSCT), is ineffective at correcting the PNS pathology. We have previously shown that systemic delivery of immortalized bone marrow-derived murine mesenchymal stromal cells (BM-MSCs) diminishes the neuropathology of transplanted Twitcher mice, a murine model of KD. In this study, to move one step closer to clinical application, the effectiveness of a systematic delivery of primary BM-MSCs to promote recovery of the Twitcher PNS was assessed. Primary BM-MSCsgrafted to the Twitcher sciatic nerve led to increased GALC activity that was not correlated to decreased psychosine (the toxic GALC substrate) accumulation. Nevertheless, BM-MSC transplantation rescued the axonal phenotype of Twitcher mice in the sciatic nerve, with an increased density of both myelinated and unmyelinated axons in transplanted animals. Whereas no increase in myelination was observed, upon transplantation an increased proliferation of Schwann cell precursors occurred. Supporting these findings, in vitro, BM-MSCs promoted neurite outgrowth of Twitcher sensory neurons and proliferation of Twitcher Schwann cells. Moreover, BM-MSCs expressed nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) and promoted increased BDNF synthesis by neighboring Schwann cells. Besides their action in neurons and glia, BM-MSCs led to macrophage activation in Twitcher sciatic nerves. In summary, primary BM-MSCs diminish the neuropathology of Twitcher sciatic nerves by coordinately affecting neurons, glia, and macrophages.
- Published
- 2014
- Full Text
- View/download PDF
15. MIQuant--semi-automation of infarct size assessment in models of cardiac ischemic injury.
- Author
-
Diana S Nascimento, Mariana Valente, Tiago Esteves, Maria de Fátima de Pina, Joana G Guedes, Ana Freire, Pedro Quelhas, and Perpétua Pinto-do-Ó
- Subjects
Medicine ,Science - Abstract
BACKGROUND: The cardiac regenerative potential of newly developed therapies is traditionally evaluated in rodent models of surgically induced myocardial ischemia. A generally accepted key parameter for determining the success of the applied therapy is the infarct size. Although regarded as a gold standard method for infarct size estimation in heart ischemia, histological planimetry is time-consuming and highly variable amongst studies. The purpose of this work is to contribute towards the standardization and simplification of infarct size assessment by providing free access to a novel semi-automated software tool. The acronym MIQuant was attributed to this application. METHODOLOGY/PRINCIPAL FINDINGS: Mice were subject to permanent coronary artery ligation and the size of chronic infarcts was estimated by area and midline-length methods using manual planimetry and with MIQuant. Repeatability and reproducibility of MIQuant scores were verified. The validation showed high correlation (r(midline length) = 0.981; r(area) = 0.970 ) and agreement (Bland-Altman analysis), free from bias for midline length and negligible bias of 1.21% to 3.72% for area quantification. Further analysis demonstrated that MIQuant reduced by 4.5-fold the time spent on the analysis and, importantly, MIQuant effectiveness is independent of user proficiency. The results indicate that MIQuant can be regarded as a better alternative to manual measurement. CONCLUSIONS: We conclude that MIQuant is a reliable and an easy-to-use software for infarct size quantification. The widespread use of MIQuant will contribute towards the standardization of infarct size assessment across studies and, therefore, to the systematization of the evaluation of cardiac regenerative potential of emerging therapies.
- Published
- 2011
- Full Text
- View/download PDF
16. Bone marrow-derived endothelial progenitors expressing Delta-like 4 (Dll4) regulate tumor angiogenesis.
- Author
-
Carla Real, Leonor Remédio, Francisco Caiado, Cátia Igreja, Cristina Borges, Alexandre Trindade, Perpétua Pinto-do-Ó, Hideo Yagita, Antonio Duarte, and Sérgio Dias
- Subjects
Medicine ,Science - Abstract
Neo-blood vessel growth (angiogenesis), which may involve the activation of pre-existing endothelial cells (EC) and/or the recruitment of bone marrow-derived vascular precursor cells (BM-VPC), is essential for tumor growth. Molecularly, besides the well established roles for Vascular endothelial growth factor (VEGF), recent findings show the Notch signalling pathway, in particular the ligand Delta-like 4 (Dll4), is also essential for adequate tumor angiogenesis; Dll4 inhibition results in impaired, non-functional, angiogenesis and reduced tumor growth. However, the role of BM-VPC in the setting of Notch pathway modulation was not addressed and is the subject of the present report. Here we show that SDF-1 and VEGF, which are produced by tumors, increase Dll4 expression on recruited BM-VPC. Mechanistically, BM-VPC activated, in a Dll4-dependent manner, a transcriptional program on mature EC suggestive of EC activation and stabilization. BM-VPC induced ICAM-2 and Fibronectin expression on EC, an effect that was blocked by a Dll4-specific neutralizing antibody. In vivo, transplantation of BM-VPC with decreased Dll4 into tumor-bearing mice resulted in the formation of microvessels with decreased pericyte coverage and reduced fibronectin expression. Consequently, transplantation of BM-VPC with decreased Dll4 resulted in impaired tumor angiogenesis, increased tumor hypoxia and apoptosis, and decreased tumor growth. Taken together, our data suggests that Dll4 expression by BM-VPC affects their communication with tumor vessel endothelial cells, thereby modulating tumor angiogenesis by affecting vascular stability.
- Published
- 2011
- Full Text
- View/download PDF
17. Learning the Biochemical Basis of Axonal Guidance: Using Caenorhabditis elegans as a Model
- Author
-
Maciel, Andreia Teixeira-Castro, João Carlos Sousa, Cármen Vieira, Joana Pereira-Sousa, Daniela Vilasboas-Campos, Fernanda Marques, Perpétua Pinto-do-Ó, and Patrícia
- Subjects
neuronal cell biology ,axon pathfinding ,growth cone ,molecular cues ,experimental activity - Abstract
Aim: Experimental models are a powerful aid in visualizing molecular phenomena. This work reports how the worm Caenorhabditis elegans (C. elegans) can be effectively explored for students to learn how molecular cues dramatically condition axonal guidance and define nervous system structure and behavior at the organism level. Summary of work: A loosely oriented observational activity preceded detailed discussions on molecules implied in axonal migration. C. elegans mutants were used to introduce second-year medical students to the deleterious effects of gene malfunctioning in neuron response to extracellular biochemical cues and to establish links between molecular function, nervous system structure, and animal behavior. Students observed C. elegans cultures and associated animal behavior alterations with the lack of function of specific axon guidance molecules (the soluble cue netrin/UNC-6 or two receptors, DCC/UNC-40 and UNC-5H). Microscopical observations of these strains, in combination with pan-neuronal GFP expression, allowed optimal visualization of severely affected neurons. Once the list of mutated genes in each strain was displayed, students could also relate abnormal patterns in axon migration/ventral and dorsal nerve cord neuron formation in C. elegans with mutated molecular components homologous to those in humans. Summary of results: Students rated the importance and effectiveness of the activity very highly. Ninety-three percent found it helpful to grasp human axonal migration, and all students were surprised with the power of the model in helping to visualize the phenomenon.
- Published
- 2023
- Full Text
- View/download PDF
18. Multiscale Analysis of Extracellular Matrix Remodeling in the Failing Heart
- Author
-
Ana Rubina Perestrelo, Vladimír Horváth, Diana S. Nascimento, Ana Catarina Silva, Kamila Koci, Alberto Rainer, Giulia Azzato, Petr Skládal, Guido Caluori, Fabiana Martino, Víta Žampachová, Giuseppe De Marco, Jorge Oliver-De La Cruz, Alessio Caravella, Ondřej Polanský, Giancarlo Forte, Perpétua Pinto-do-Ó, Vladimír Vinarský, and Stefania Pagliari
- Subjects
Cardiomyopathy, Dilated ,Physiology ,Myocardial Infarction ,Failing heart ,030204 cardiovascular system & hematology ,Mechanotransduction, Cellular ,Ventricular Function, Left ,Cardiac cell ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Animals ,Humans ,Elasticity (economics) ,Cells, Cultured ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,Heart Failure ,0303 health sciences ,Ventricular Remodeling ,Chemistry ,Myocardium ,YAP-Signaling Proteins ,Fibroblasts ,Extracellular Matrix ,Cell biology ,Mice, Inbred C57BL ,Disease Models, Animal ,Molecular network ,Case-Control Studies ,Cardiology and Cardiovascular Medicine ,Transcription Factors - Abstract
Rationale: Cardiac ECM (extracellular matrix) comprises a dynamic molecular network providing structural support to heart tissue function. Understanding the impact of ECM remodeling on cardiac cells during heart failure (HF) is essential to prevent adverse ventricular remodeling and restore organ functionality in affected patients. Objectives: We aimed to (1) identify consistent modifications to cardiac ECM structure and mechanics that contribute to HF and (2) determine the underlying molecular mechanisms. Methods and Results: We first performed decellularization of human and murine ECM (decellularized ECM) and then analyzed the pathological changes occurring in decellularized ECM during HF by atomic force microscopy, 2-photon microscopy, high-resolution 3-dimensional image analysis, and computational fluid dynamics simulation. We then performed molecular and functional assays in patient-derived cardiac fibroblasts based on YAP (yes-associated protein)-transcriptional enhanced associate domain (TEAD) mechanosensing activity and collagen contraction assays. The analysis of HF decellularized ECM resulting from ischemic or dilated cardiomyopathy, as well as from mouse infarcted tissue, identified a common pattern of modifications in their 3-dimensional topography. As compared with healthy heart, HF ECM exhibited aligned, flat, and compact fiber bundles, with reduced elasticity and organizational complexity. At the molecular level, RNA sequencing of HF cardiac fibroblasts highlighted the overrepresentation of dysregulated genes involved in ECM organization, or being connected to TGFβ1 (transforming growth factor β1), interleukin-1, TNF-α, and BDNF signaling pathways. Functional tests performed on HF cardiac fibroblasts pointed at mechanosensor YAP as a key player in ECM remodeling in the diseased heart via transcriptional activation of focal adhesion assembly. Finally, in vitro experiments clarified pathological cardiac ECM prevents cell homing, thus providing further hints to identify a possible window of action for cell therapy in cardiac diseases. Conclusions: Our multiparametric approach has highlighted repercussions of ECM remodeling on cell homing, cardiac fibroblast activation, and focal adhesion protein expression via hyperactivated YAP signaling during HF.
- Published
- 2021
- Full Text
- View/download PDF
19. Myocardial Edema: an Overlooked Mechanism of Septic Cardiomyopathy?
- Author
-
Adelino F. Leite-Moreira, Roberto Roncon-Albuquerque, José A. Paiva, Nuno Bettencourt, Perpétua Pinto-do-Ó, Francisco Vasques-Nóvoa, António J. Madureira, Fátima Carneiro, Tiago L. Laundos, Diana S. Nascimento, and José Pedro L. Nunes
- Subjects
Adult ,Male ,medicine.medical_specialty ,Myocardial edema ,030204 cardiovascular system & hematology ,Critical Care and Intensive Care Medicine ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Edema ,Humans ,Medicine ,Septic cardiomyopathy ,business.industry ,Mechanism (biology) ,Septic shock ,030208 emergency & critical care medicine ,Middle Aged ,bacterial infections and mycoses ,medicine.disease ,Shock, Septic ,Pathophysiology ,Emergency Medicine ,Cardiology ,Female ,Cardiomyopathies ,business - Abstract
Septic cardiomyopathy is an increasingly relevant topic in clinical management of septic shock. However, pathophysiological mechanisms and long-term consequences of sepsis-induced myocardial injury are still poorly understood. Herein, new clinical and histological evidence is provided suggesting an association of myocardial edema formation with tissue injury and subsequent remodeling in septic shock patients. This preliminary data supports myocardial edema as a potentially relevant and largely unexplored mechanism of human septic cardiomyopathy.
- Published
- 2020
- Full Text
- View/download PDF
20. Proteomic Identification of a Gastric Tumor ECM Signature Associated With Cancer Progression
- Author
-
Ana M. Moreira, Rui M. Ferreira, Patrícia Carneiro, Joana Figueiredo, Hugo Osório, José Barbosa, John Preto, Perpétua Pinto-do-Ó, Fátima Carneiro, and Raquel Seruca
- Subjects
Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Molecular Biology ,Biochemistry - Abstract
The extracellular matrix (ECM) plays an undisputable role in tissue homeostasis and its deregulation leads to altered mechanical and biochemical cues that impact cancer development and progression. Herein, we undertook a novel approach to address the role of gastric ECM in tumorigenesis, which remained largely unexplored. By combining decellularization techniques with a high-throughput quantitative proteomics approach, we have performed an extensive characterization of human gastric mucosa, uncovering its composition and distribution among tumor, normal adjacent and normal distant mucosa. Our results revealed a common ECM signature composed of 142 proteins and indicated that gastric carcinogenesis encompasses ECM remodeling through alterations in the abundance of 24 components, mainly basement membrane proteins. Indeed, we could only identify one de novo tumor-specific protein, the collagen alpha-1(X) chain (COL10A1). Functional analysis of the data demonstrated that gastric ECM remodeling favors tumor progression by activating ECM receptors and cellular processes involved in angiogenesis and cell-extrinsic metabolic regulation. By analyzing mRNA expression in an independent GC cohort available at the TGCA, we validated the expression profile of 12 differentially expressed ECM proteins. Importantly, the expression of COL1A2, LOX and LTBP2 significantly correlated with high tumor stage, with LOX and LTBP2 further impacting patient overall survival. These findings contribute for a better understanding of GC biology and highlight the role of core ECM components in gastric carcinogenesis and their clinical relevance as biomarkers of disease prognosis.
- Published
- 2021
21. Pharmacological clearance of senescent cells reverses HFpEF hallmarks by decreasing inflammation, endothelial dysfunction and cardiac fibrosis
- Author
-
Adelino F. Leite-Moreira, Alexandre Ortega Gonçalves, Inês Falcão-Pires, Lino Ferreira, António Barros, Perpétua Pinto-do-Ó, Daniel Sousa, Susana G. Santos, Nádia Gonçalves, Andréia Aparecida da Silva, António Angélico-Gonçalves, Carolina Caetano, Patrícia R. Pitrez, Diana S. Nascimento, Glória Conceição, Elsa D. Silva, Francisco Vasques-Nóvoa, Inês Tomé, Daniela Miranda-Silva, and Vasco Sampaio-Pinto
- Subjects
Pathology ,medicine.medical_specialty ,Text mining ,business.industry ,Cardiac fibrosis ,medicine ,Inflammation ,Endothelial dysfunction ,medicine.symptom ,medicine.disease ,business - Abstract
Aging and chronic inflammation are associated with the development of heart failure with preserved ejection fraction (HFpEF). However, cellular senescence as a potential mechanistic link between both events and its pathophysiological and therapeutic role were yet unexplored. Here we show that ZSF1-obese rats, a model of cardiometabolic HFpEF, have exacerbated systemic inflammation and endothelial damage compared to ZSF1-lean littermates. In addition, ZSF1-obese rats accumulated immune and endothelial senescent cells in the peripheral blood and myocardium. Accordingly, the frequency of circulating senescent leukocytes associated with markers of disease severity in HFpEF patients. Notably, systemic treatment of ZSF1-obese rats with Navitoclax, a BCL-2 family inhibitor, reduced senescent cell burden, decreased circulating B-type natriuretic peptide levels, and attenuated inflammation, vascular remodeling and cardiac fibrosis. Our findings advance cellular senescence as a key mechanistic pathway leading to HFpEF and provide proof-of-concept evidence that senolytics are a promising treatment for this disease.
- Published
- 2021
- Full Text
- View/download PDF
22. Yolk sac, but not hematopoietic stem cell–derived progenitors, sustain erythropoiesis throughout murine embryonic life
- Author
-
Francisca Soares-da-Silva, Laina Freyer, Ramy Elsaid, Odile Burlen-Defranoux, Lorea Iturri, Odile Sismeiro, Perpétua Pinto-do-Ó, Elisa Gomez-Perdiguero, Ana Cumano
- Published
- 2021
- Full Text
- View/download PDF
23. Yolk sac erythromyeloid progenitors sustain erythropoiesis throughout embryonic life
- Author
-
Francisca Soares-da-Silva, Laina Freyer, Ana Cumano, Odile Sismeiro, Lorea Iturri, Ramy Elsaid, Elisa Gomez-Perdiguero, Odile Burlen-Defranoux, and Perpétua Pinto-do-Ó
- Subjects
0303 health sciences ,Hematopoietic stem cell ,Biology ,Embryonic stem cell ,Cell biology ,03 medical and health sciences ,Haematopoiesis ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,embryonic structures ,medicine ,Erythropoiesis ,Bone marrow ,Progenitor cell ,Yolk sac ,Stem cell ,030304 developmental biology - Abstract
The first hematopoietic cells are produced in the yolk sac and are thought to be rapidly replaced by the progeny of hematopoietic stem cells. Here we document that hematopoietic stem cells do not contribute significantly to erythrocyte production up until birth. Lineage tracing of yolk sac-derived erythromyeloid progenitors, that also contribute to tissue resident macrophages, shows a progeny of highly proliferative erythroblasts, that after intra embryonic injection, rapidly differentiate. These progenitors, similar to hematopoietic stem cells, arec-Mybdependent and are developmentally restricted as they are not found in the bone marrow. We show that erythrocyte progenitors of yolk sac origin require lower concentrations of erythropoietin than their hematopoietic stem cell-derived counterparts for efficient erythrocyte production. Consequently, fetal liver hematopoietic stem cells fail to generate megakaryocyte and erythrocyte progenitors. We propose that large numbers of yolk sac-derived erythrocyte progenitors have a selective advantage and efficiently outcompete hematopoietic stem cell progeny in an environment with limited availability of erythropoietin.
- Published
- 2020
- Full Text
- View/download PDF
24. Neonatal Apex Resection Triggers Cardiomyocyte Proliferation, Neovascularization and Functional Recovery Despite Local Fibrosis
- Author
-
Perpétua Pinto-do-Ó, Ana Catarina Silva, Rui J Cerqueira, Francisco Vasques-Nóvoa, Adelino F. Leite-Moreira, Elsa D. Silva, Diana S. Nascimento, Gabriele D'Uva, Sílvia C. Rodrigues, Nicola Pianca, Tiago L. Laundos, Sólveig Thorsteinsdóttir, Tatiana P. Resende, Vasco Sampaio-Pinto, Instituto de Investigação e Inovação em Saúde, Sampaio-Pinto V., Rodrigues S.C., Laundos T.L., Silva E.D., Vasques-Novoa F., Silva A.C., Cerqueira R.J., Resende T.P., Pianca N., Leite-Moreira A., D'Uva G., Thorsteinsdottir S., Pinto-do-O P., and Nascimento D.S.
- Subjects
0301 basic medicine ,Fibrosi ,cardiac injury response ,Biochemistry ,Heart Ventricle ,Neovascularization ,Extracellular matrix ,Mice ,0302 clinical medicine ,Heart Ventricles / physiopathology ,Fibrosis ,Myocytes, Cardiac ,Cardiomyocyte proliferation ,lcsh:QH301-705.5 ,cardiac fibroblasts ,lcsh:R5-920 ,Neovascularization, Pathologic ,Neovascularization, Pathologic / physiopathology ,cardiac regeneration ,Fibrosis / physiopathology ,Heart ,Pathophysiology ,Myocytes, Cardiac / physiology ,cardiomyocyte proliferation ,Cardiology ,Myocardium / pathology ,cardiovascular system ,medicine.symptom ,neovascularization ,Heart Injuries / physiopathology ,lcsh:Medicine (General) ,Heart Injurie ,medicine.medical_specialty ,Heart Injury ,Heart Ventricles ,extracellular matrix ,Biology ,Article ,03 medical and health sciences ,Internal medicine ,Genetics ,medicine ,Animals ,Regeneration ,Heart / physiology ,Cell Proliferation ,Animal ,Regeneration (biology) ,Myocardium ,fibrosis ,cardiac fibroblast ,Cell Biology ,Recovery of Function ,medicine.disease ,Cell Proliferation / physiology ,Apex (geometry) ,Mice, Inbred C57BL ,030104 developmental biology ,neonatal apex resection ,Animals, Newborn ,Heart Injuries ,lcsh:Biology (General) ,Regeneration / physiology ,stereology ,030217 neurology & neurosurgery ,Recovery of Function / physiology ,Developmental Biology - Abstract
Summary So far, opposing outcomes have been reported following neonatal apex resection in mice, questioning the validity of this injury model to investigate regenerative mechanisms. We performed a systematic evaluation, up to 180 days after surgery, of the pathophysiological events activated upon apex resection. In response to cardiac injury, we observed increased cardiomyocyte proliferation in remote and apex regions, neovascularization, and local fibrosis. In adulthood, resected hearts remain consistently shorter and display permanent fibrotic tissue deposition in the center of the resection plane, indicating limited apex regrowth. However, thickening of the left ventricle wall, explained by an upsurge in cardiomyocyte proliferation during the initial response to injury, compensated cardiomyocyte loss and supported normal systolic function. Thus, apex resection triggers both regenerative and reparative mechanisms, endorsing this injury model for studies aimed at promoting cardiomyocyte proliferation and/or downplaying fibrosis., Graphical Abstract, Highlights • Apex resection triggers fibrosis, neovascularization, and cardiomyocyte proliferation • Permanent fibrotic deposition is confined to the apex • Injured hearts display morphometric alterations but regain functional competence • Cardiomyocyte proliferation is sufficient to compensate tissue loss by resection, In this article, Nascimento and colleagues demonstrate that neonatal apex resection stimulates cardiomyocyte proliferation and permanent scarring in the apex. Newly formed cardiomyocytes compensate muscle loss by resection, and resected hearts recover functional competence in adulthood. These findings endorse this model for studies aiming to block cardiac fibrosis and/or favoring CM proliferation.
- Published
- 2018
25. Transient HES5 Activity Instructs Mesodermal Cells toward a Cardiac Fate
- Author
-
Carlos Filipe Pereira, Francisca Soares-da-Silva, Tatiana P. Resende, Dung Fang Lee, Avinash Waghray, Ana G. Freire, Ihor R. Lemischka, Diana S. Nascimento, and Perpétua Pinto-do-Ó
- Subjects
0301 basic medicine ,Mesoderm ,Cellular differentiation ,notch signaling pathway ,Regulator ,Notch signaling pathway ,HES5 ,Biology ,Biochemistry ,Article ,Cell Line ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Genetics ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Erythropoiesis ,Myocytes, Cardiac ,lcsh:QH301-705.5 ,Cell Proliferation ,lcsh:R5-920 ,Gastrulation ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Mouse Embryonic Stem Cells ,Cell Biology ,Embryonic stem cell ,Cell biology ,Repressor Proteins ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,nascent mesoderm ,Gene Knockdown Techniques ,ISL1 ,Cancer research ,cardiac fate specification ,lcsh:Medicine (General) ,Hes5 ,030217 neurology & neurosurgery ,Developmental Biology ,Signal Transduction - Abstract
Summary Notch signaling plays a role in specifying a cardiac fate but the downstream effectors remain unknown. In this study we implicate the Notch downstream effector HES5 in cardiogenesis. We show transient Hes5 expression in early mesoderm of gastrulating embryos and demonstrate, by loss and gain-of-function experiments in mouse embryonic stem cells, that HES5 favors cardiac over primitive erythroid fate. Hes5 overexpression promotes upregulation of the cardiac gene Isl1, while the hematopoietic regulator Scl is downregulated. Moreover, whereas a pulse of Hes5 instructs cardiac commitment, sustained expression after lineage specification impairs progression of differentiation to contracting cardiomyocytes. These findings establish a role for HES5 in cardiogenesis and provide insights into the early cardiac molecular network., Graphical Abstract, Highlights • Hes5 is expressed in the nascent mesoderm of gastrulating mouse embryos • Hes5 knockdown enhances primitive erythropoiesis in mESCs • A stage-specific pulse of Hes5 instructs preferential cardiac fate in mESCs • Sustained Hes5 activation impairs differentiation to contracting cardiomyocytes, Pinto-do-Ó and colleagues establish a role for HES5 in instructing cardiac versus primitive erythroid fate. Hes5 depletion enhances primitive erythropoiesis, whereas a stage-specific overexpression favors cardiac specification in mESCs. Progression of differentiation to contracting cardiomyocytes depends on Hes5 downregulation. This requirement for transient Hes5 activity for proper cardiogenesis correlates with the expression pattern observed in the mouse embryo nascent mesoderm.
- Published
- 2017
26. Restoring heart function and electrical integrity: closing the circuit
- Author
-
Francisco Vasques-Nóvoa, Luís Miguel Monteiro, Lino Ferreira, Perpétua Pinto-do-Ó, and Diana S. Nascimento
- Subjects
0301 basic medicine ,Future studies ,Heart disease ,media_common.quotation_subject ,Biomedical Engineering ,Medicine (miscellaneous) ,Physiology ,Review Article ,030204 cardiovascular system & hematology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Electrical integrity ,Function (engineering) ,media_common ,business.industry ,Cell Biology ,medicine.disease ,Cell delivery ,3. Good health ,030104 developmental biology ,Medicine ,Electrical conduction system of the heart ,business ,Neuroscience ,Developmental Biology - Abstract
Cardiovascular diseases are the main cause of death in the world and are often associated with the occurrence of arrhythmias due to disruption of myocardial electrical integrity. Pathologies involving dysfunction of the specialized cardiac excitatory/conductive tissue are also common and constitute an added source of morbidity and mortality since current standard therapies withstand a great number of limitations. As electrical integrity is essential for a well-functioning heart, innovative strategies have been bioengineered to improve heart conduction and/or promote myocardial repair, based on: (1) gene and/or cell delivery; or (2) conductive biomaterials as tools for cardiac tissue engineering. Herein we aim to review the state-of-art in the area, while briefly describing the biological principles underlying the heart electrical/conduction system and how this system can be disrupted in heart disease. Suggestions regarding targets for future studies are also presented.
- Published
- 2017
27. HSA+immature cardiomyocytes persist in the adult heart and expand after ischemic injury
- Author
-
Mariana Valente, Ana Cumano, Benoit Dupont, Odile Burlen-Defranoux, Tatiana P. Resende, Perpétua Pinto-do-Ó, and Diana S. Nascimento
- Subjects
0303 health sciences ,Fetus ,education.field_of_study ,CD24 ,Cell ,Population ,Infarction ,Biology ,medicine.disease ,Cell biology ,Transplantation ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,In vivo ,medicine ,Progenitor cell ,education ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
The assessment of the regenerative capacity of the heart has been compromised by the lack of surface signatures to characterize cardiomyocytes. Here, combined multiparametric surface marker analysis with single cell transcriptional profiling and in vivo transplantation, identify the main fetal cardiac populations and their progenitors. We found that cardiomyocytes at different stages of differentiation co-exist during development. We identified a population of immature HSA/CD24+cardiomyocytes that persists throughout life and that, unlike other cardiomyocyte subsets, actively proliferates up to one week of age and engraft cardiac tissue upon transplantation. In adult heart HSA/CD24+cardiomyocytes appear as mononucleated cells that cycle and increase in frequency after infarction. Our work identified cell surface signatures that allow the prospective isolation of cardiomyocytes at any developmental stage and the detection of adult cardiomyocytes poised for activation in response to ischemic stimuli. This work opens new perspectives in the understanding and treatment of heart pathologies.
- Published
- 2019
- Full Text
- View/download PDF
28. Establishing a link between endothelial cell metabolism and vascular behaviour in a type 1 diabetes mouse model
- Author
-
Raquel Soares, Sara Andrade, Cláudia G. Silva, Eugenia Carvalho, Sampaio-Pinto, Ilda Rodrigues, Diana S. Nascimento, Raquel Costa, Perpétua Pinto-do-Ó, Susana G. Guerreiro, and Instituto de Investigação e Inovação em Saúde
- Subjects
0301 basic medicine ,Male ,Pathology ,Physiology ,Angiogenesis ,medicine.medical_treatment ,Connective Tissue Growth Factor / analysis ,Carbohydrate and lipid metabolism ,Kidney / cytology ,Microvessels / pathology ,Kidney ,lcsh:Physiology ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Fibrosis ,Transforming Growth Factor beta ,Transforming Growth Factor beta / metabolism ,lcsh:QD415-436 ,Macrovascular complications ,Vascular Endothelial Growth Factor Receptor-2 / genetics ,Tissue Inhibitor of Metalloproteinase-2 / genetics ,lcsh:QP1-981 ,Receptors, Notch ,Neovascularization, Pathologic ,Endothelial Cells / metabolism ,Genomics ,Platelet Endothelial Cell Adhesion Molecule-1 / metabolism ,3. Good health ,Endothelial stem cell ,Platelet Endothelial Cell Adhesion Molecule-1 ,Cell sorting ,Diabetes Mellitus, Experimental / pathology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Diabetes Mellitus, Experimental / chemically induced ,Receptors, Notch / metabolism ,Diabetes Mellitus, Experimental / metabolism ,Endothelial Cells / cytology ,Kidney / metabolism ,Microvessels / physiology ,medicine.medical_specialty ,Myocardium / metabolism ,Heart Ventricles ,Micro and macrovascular complications ,Transforming Growth Factor beta / genetics ,Article ,Diabetes Mellitus, Experimental ,lcsh:Biochemistry ,03 medical and health sciences ,Connective Tissue Growth Factor / metabolism ,Diabetes mellitus ,medicine ,Vascular Endothelial Growth Factor Receptor-2 / metabolism ,Animals ,Heart Ventricles / metabolism ,Tissue Inhibitor of Metalloproteinase-2 / metabolism ,Sirius Red ,Tissue Inhibitor of Metalloproteinase-2 ,business.industry ,Growth factor ,Myocardium ,Connective Tissue Growth Factor ,Micro ,Endothelial Cells ,medicine.disease ,Vascular Endothelial Growth Factor Receptor-2 ,CTGF ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Endothelium metabolism ,Microvessels ,business ,Myocardium / cytology ,Transcriptome - Abstract
Background/Aims: Vascular complications contribute significantly to the extensive morbidity and mortality rates observed in people with diabetes. Despite well known that the diabetic kidney and heart exhibit imbalanced angiogenesis, the mechanisms implicated in this angiogenic paradox remain unknown. In this study, we examined the angiogenic and metabolic gene expression profile (GEP) of endothelial cells (ECs) isolated from a mouse model with type1 diabetes mellitus (T1DM). Methods: ECs were isolated from kidneys and hearts of healthy and streptozocin (STZ)-treated mice. RNA was then extracted for molecular studies. GEP of 84 angiogenic and 84 AMP-activated Protein Kinase (AMPK)-dependent genes were examined by microarrays. Real time PCR confirmed the changes observed in significantly altered genes. Microvessel density (MVD) was analysed by immunohistochemistry, fibrosis was assessed by the Sirius red histological staining and connective tissue growth factor (CTGF) was quantified by ELISA. Results: The relative percentage of ECs and MVD were increased in the kidneys of T1DM animals whereas the opposite trend was observed in the hearts of diabetic mice. Accordingly, the majority of AMPK-associated genes were upregulated in kidneys and downregulated in hearts of these animals. Angiogenic GEP revealed significant differences in Tgfß, Notch signaling and Timp2 in both diabetic organs. These findings were in agreement with the angiogenesis histological assays. Fibrosis was augmented in both organs in diabetic as compared to healthy animals. Conclusion: Altogether, our findings indicate, for the first time, that T1DM heart and kidney ECs present opposite metabolic cues, which are accompanied by distinct angiogenic patterns. These findings enable the development of innovative organ-specific therapeutic strategies targeting diabetic-associated vascular disorders. This work was supported by CAPES (Sciences without Borders - Full Doctorate Fellowship – Process 10010-13-0); FEDER funds by COMPETE: [POCI-01-0145-FEDER-007440, POCI-01-0145-FEDER-016385]; NORTE2020 [NORTE-01-0145FEDER-000012]; HealthyAging2020 [CENTRO-01-0145-FEDER-000012-N2323]; FCT - Fundação para a Ciência e a Tecnologia [UID/BIM/04293/2013, EXPL/BIM-MED/0492/2012, SFRH/BPD/88745/2012, SFRH/BD/111799/2015]; Claude Pepper Older Americans Independence Center; grant: P30 AG028718, NIGMS Award P20GM109096; European Structural and Investment Funds (ESIF). AUTHOR CONTRIBUTION: CS and RS participated in the design and conception of the study; CS performed the whole laboratory and statistical analyses and drafted the manuscript; VSP, PPO, DSN carried out the FACS assay design and data acquisition, as well as the interpretation of FACS data; SA advised and performed microarray and RT-PCR assays; IR headed the parafin embedded tissue and histologial staining; SG, EC were responsible for the animal studies and immunohistochemistry analyses; RC advised the methodological laboratorial analysis and animal studies; RS and EC critically revised the manuscript for important intellectual content. All authors were involved in drafting and revising the article. All authors read and approved the final version of the manuscript.
- Published
- 2019
29. Decellularized human colorectal cancer matrices polarize macrophages towards an anti-inflammatory phenotype promoting cancer cell invasion via CCL18
- Author
-
Ana Pinto, Mário A. Barbosa, Perpétua Pinto-do-Ó, Ana C. Silva, Elisabete Rios, A.P. Cardoso, Cristina C. Barrias, Nuno C. Santos, Maria José Oliveira, Fátima Carneiro, Cecília Durães, Filomena A. Carvalho, Marta Pinto, Hugo R. Caires, Sara C. Neves, Diana S. Nascimento, and Repositório da Universidade de Lisboa
- Subjects
0301 basic medicine ,Colorectal cancer ,Cell ,Biophysics ,Macrophage polarization ,Bioengineering ,Biology ,Biomaterials ,Extracellular matrix ,03 medical and health sciences ,medicine ,Tumor Cells, Cultured ,Tumor Microenvironment ,CCL18 ,Humans ,Neoplasm Invasiveness ,Decellularization ,Cell-Free System ,Tissue Scaffolds ,Macrophages ,Cancer ,Cell Polarity ,medicine.disease ,3. Good health ,Extracellular Matrix ,030104 developmental biology ,medicine.anatomical_structure ,Mechanics of Materials ,Chemokines, CC ,Cancer cell ,Immunology ,Ceramics and Composites ,Cancer research ,Tumor necrosis factor alpha ,Colorectal Neoplasms - Abstract
© 2017 Elsevier Ltd, Macrophages are frequently identified in solid tumors, playing important roles in cancer progression. Their remarkable plasticity makes them very sensitive to environmental factors, including the extracellular matrix (ECM). In the present work, we investigated the impact of human colorectal tumor matrices on macrophage polarization and on macrophage-mediated cancer cell invasion. Accordingly, we developed an innovative 3D-organotypic model, based on the decellularization of normal and tumor tissues derived from colorectal cancer patients' surgical resections. Extensive characterization of these scaffolds revealed that DNA and other cell constituents were efficiently removed, while native tissue characteristics, namely major ECM components, architecture and mechanical properties, were preserved. Notably, normal and tumor decellularized matrices distinctly promoted macrophage polarization, with macrophages in tumor matrices differentiating towards an anti-inflammatory M2-like phenotype (higher IL-10, TGF-β and CCL18 and lower CCR7 and TNF expression). Matrigel invasion assays revealed that tumor ECM-educated macrophages efficiently stimulated cancer cell invasion through a mechanism involving CCL18. Notably, the high expression of this chemokine at the invasive front of human colorectal tumors correlated with advanced tumor staging. Our approach evidences that normal and tumor decellularized matrices constitute excellent scaffolds when trying to recreate complex microenvironments to understand basic mechanisms of disease or therapeutic resistance., This work was financially supported by FEDER - Fundo Europeu de Desenvolvimento Regional funds through the COMPETE 2020 - Operacional Programme for Competitiveness and Internationalisation (POCI), Portugal 2020, and by Portuguese funds through FCT - Fundação para a Ciência e a Tecnologia/Ministério da Ciência, Tecnologia e Inovaçao in the framework of the project “Institute for Research and Innovation in Health Sciences” (POCI-01-0145-FEDER-007274), FCT2012-Investigator Program (IF/01066/2012 Maria Jose Oliveira), PhD fellowships (SFRH/BD/81103/2011, SFRH/BD/88780/2012, SFRH/BD/76995/2011 and SFRH/BD/74144/2010) and Postdoctoral fellowship (SFRH/BPD/99442/2014). We also would like to thank Dina Leitão (i3S/IPATIMUP, Porto, Portugal) for her assistance with the CCL18 immunohistochemistry and a special thanks to Daniela Silva (CEMUP, Porto, Portugal) for the SEM analysis.
- Published
- 2017
30. Exosomes secreted by cardiomyocytes subjected to ischaemia promote cardiac angiogenesis
- Author
-
Brenda R. Kwak, Vanessa Coelho-Santos, Marta Pinto, Daniela Batista-Almeida, Mónica Zuzarte, Paulo Pereira, Henrique Girão, Diana S. Nascimento, Lino Ferreira, Ricardo Pereira, Rosa Fernandes, Francisco J. Enguita, Marina C. Costa, Ana P. Silva, Pedro Gouveia, Rita Pereira-Carvalho, Perpétua Pinto-do-Ó, Teresa Ribeiro-Rodrigues, Justin C Mason, Tiago L. Laundos, and Repositório da Universidade de Lisboa
- Subjects
0301 basic medicine ,Physiology ,Angiogenesis ,Ischemia ,ddc:616.07 ,Coronary collateral circulation ,Exosomes ,Ischaemia ,Exosome ,Neovascularization ,03 medical and health sciences ,Cell Movement ,Physiology (medical) ,Morphogenesis ,Medicine ,Animals ,Myocytes, Cardiac ,Rats, Wistar ,Cells, Cultured ,Cardiac muscle cell ,Matrigel ,Neovascularization, Pathologic ,business.industry ,Myocardium ,Endothelial Cells ,Cell migration ,Biological Transport ,Extracellular vesicles ,medicine.disease ,Microvesicles ,3. Good health ,Cell biology ,Myocardial infarction ,030104 developmental biology ,medicine.anatomical_structure ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business - Abstract
Copyright © 2017, Oxford University Press, Aims: Myocardial infarction (MI) is the leading cause of morbidity and mortality worldwide and results from an obstruction in the blood supply to a region of the heart. In an attempt to replenish oxygen and nutrients to the deprived area, affected cells release signals to promote the development of new vessels and confer protection against MI. However, the mechanisms underlying the growth of new vessels in an ischaemic scenario remain poorly understood. Here, we show that cardiomyocytes subjected to ischaemia release exosomes that elicit an angiogenic response of endothelial cells (ECs). Methods and results: Exosomes secreted by H9c2 myocardial cells and primary cardiomyocytes, cultured either in control or ischaemic conditions were isolated and added to ECs. We show that ischaemic exosomes, in comparison with control exosomes, confer protection against oxidative-induced lesion, promote proliferation, and sprouting of ECs, stimulate the formation of capillary-like structures and strengthen adhesion complexes and barrier properties. Moreover, ischaemic exosomes display higher levels of metalloproteases (MMP) and promote the secretion of MMP by ECs. We demonstrate that miR-222 and miR-143, the relatively most abundant miRs in ischaemic exosomes, partially recapitulate the angiogenic effect of exosomes. Additionally, we show that ischaemic exosomes stimulate the formation of new functional vessels in vivo using in ovo and Matrigel plug assays. Finally, we demonstrate that intramyocardial delivery of ischaemic exosomes improves neovascularization following MI. Conclusions: This study establishes that exosomes secreted by cardiomyocytes under ischaemic conditions promote heart angiogenesis, which may pave the way towards the development of add-on therapies to enhance myocardial blood supply., This work was supported by European Regional Development Fund (FEDER) through the Operational Program for Competitiveness Factors (COMPETE) [HealthyAging2020 CENTRO-01-0145-FEDER-000012-N2323, POCI-01-0145-FEDER-016385, POCI-01-0145-FEDER-007440 to CNC.IBILI, POCI-01-0145-FEDER-007274 to i3S/INEB and NORTE-01-0145- FEDER-000012 to T.L.L.]; national funds through the Portuguese Foundation for Science and Technology (FCT) [PTDC/SAU-ORG/119296/2010, PTDC/NEU-OSD/0312/2012, PESTC/ SAU/UI3282/2013-2014, MITP-TB/ECE/0013/2013, FCT-UID/NEU/04539/2013], PD/BD/52294/2013 to T.M.R.R., SFRH/BD/85556/2012 (co-financed by QREN) to V.C.S]; Lisboa Portugal Regional Operational Programme (LISBOA 2020) and Norte Portugal Regional Operational Programme (NORTE 2020), under the PORTUGAL 2020 Partnership Agreement; and by INFARMED Autoridade Nacional do Medicamento e Produtos de Saúde, I.P. [FIS-FIS-2015-01_CCV_20150630-157].
- Published
- 2017
31. Primary Bone Marrow Mesenchymal Stromal Cells Rescue the Axonal Phenotype of Twitcher Mice
- Author
-
Vera Sousa, Mónica Mendes Sousa, Ana Marques, Márcia A. Liz, Carla Teixeira, Telma Emanuela Santos, Perpétua Pinto-do-Ó, and Catarina Oliveira Miranda
- Subjects
Male ,Neurite ,Green Fluorescent Proteins ,Biomedical Engineering ,lcsh:Medicine ,Schwann cell ,Enzyme-Linked Immunosorbent Assay ,Mice, Transgenic ,Neurotrophic factors ,Nerve Growth Factor ,medicine ,Animals ,Cells, Cultured ,Transplantation ,Reverse Transcriptase Polymerase Chain Reaction ,business.industry ,Brain-Derived Neurotrophic Factor ,lcsh:R ,Mesenchymal stem cell ,Psychosine ,Mesenchymal Stem Cells ,Cell Biology ,Cell biology ,Nerve growth factor ,medicine.anatomical_structure ,nervous system ,Peripheral nervous system ,Immunology ,Female ,Sciatic nerve ,business - Abstract
Krabbe's disease (KD) is a demyelinating disorder caused by the deficiency of lysosomal galactocerebrosi-dase (GALC), affecting both the central (CNS) and the peripheral nervous system (PNS). A current therapy, hematopoietic stem cell transplantation (HSCT), is ineffective at correcting the PNS pathology. We have previously shown that systemic delivery of immortalized bone marrow-derived murine mesenchymal stromal cells (BM-MSCs) diminishes the neuropathology of transplanted Twitcher mice, a murine model of KD. In this study, to move one step closer to clinical application, the effectiveness of a systematic delivery of primary BM-MSCs to promote recovery of the Twitcher PNS was assessed. Primary BM-MSCsgrafted to the Twitcher sciatic nerve led to increased GALC activity that was not correlated to decreased psychosine (the toxic GALC substrate) accumulation. Nevertheless, BM-MSC transplantation rescued the axonal phenotype of Twitcher mice in the sciatic nerve, with an increased density of both myelinated and unmyelinated axons in transplanted animals. Whereas no increase in myelination was observed, upon transplantation an increased proliferation of Schwann cell precursors occurred. Supporting these findings, in vitro, BM-MSCs promoted neurite outgrowth of Twitcher sensory neurons and proliferation of Twitcher Schwann cells. Moreover, BM-MSCs expressed nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) and promoted increased BDNF synthesis by neighboring Schwann cells. Besides their action in neurons and glia, BM-MSCs led to macrophage activation in Twitcher sciatic nerves. In summary, primary BM-MSCs diminish the neuropathology of Twitcher sciatic nerves by coordinately affecting neurons, glia, and macrophages.
- Published
- 2014
- Full Text
- View/download PDF
32. Mouse HSA+ immature cardiomyocytes persist in the adult heart and expand after ischemic injury
- Author
-
Diana S. Nascimento, Benoit Dupont, Odile Burlen-Defranoux, Francisca Soares-da-Silva, Tatiana P. Resende, Mariana Valente, Ana Cumano, Perpétua Pinto-do-Ó, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Instituto de Investigação e Inovação em Saúde (I3S), Instituto de Engenharia Biomédica (INEB), Lymphopoïèse (Lymphopoïèse (UMR_1223 / U1223 / U-Pasteur_4)), Institut Pasteur [Paris]-Université Paris Diderot - Paris 7 (UPD7)-Institut National de la Santé et de la Recherche Médicale (INSERM), Cellule Pasteur, Université Paris Diderot - Paris 7 (UPD7)-PRES Sorbonne Paris Cité, Beckman Coulter Genom SA, This work was financed by European Structural and Investment Funds (ESIF), under Lisbon Portugal Regional Operational Program and National Funds through FCT-Foundation for Science and Technology under project POCI-01-0145-FEDER-016385 to PPO, by Pasteur Institute, INSERM, ANR (grant Twothyme), REVIVE Future Investment Program and Pasteur-Weizmann Foundation through grants to AC. MV (SFRH/BD/74218/2010) and TPR (SFRH/BPD/80588/2011) were supported by FCT, and PPO was recipient of an invited scientist grant by Institut Pasteur, Paris, France. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript., ANR-14-CE11-0022,Twothyme,Deux progéniteurs hématopoïétiques différents établissent le compartiment de lymphocytes T: tester un nouveau paradigme du développement T.(2014), ANR-10-LABX-0073,REVIVE,Stem Cells in Regenerative Biology and Medicine(2010), Instituto de Investigação e Inovação em Saúde, Universidade do Porto = University of Porto, Institut Pasteur [Paris] (IP)-Université Paris Diderot - Paris 7 (UPD7)-Institut National de la Santé et de la Recherche Médicale (INSERM), Vougny, Marie-Christine, Appel à projets générique - Deux progéniteurs hématopoïétiques différents établissent le compartiment de lymphocytes T: tester un nouveau paradigme du développement T. - - Twothyme2014 - ANR-14-CE11-0022 - Appel à projets générique - VALID, and Laboratoires d'excellence - Stem Cells in Regenerative Biology and Medicine - - REVIVE2010 - ANR-10-LABX-0073 - LABX - VALID
- Subjects
Male ,0301 basic medicine ,Physiology ,Cellular differentiation ,Myocardial Ischemia ,Gene Expression ,Cell Lineage / physiology ,Biochemistry ,Myocytes, Cardiac / metabolism ,Mice ,Spectrum Analysis Techniques ,0302 clinical medicine ,Single-cell analysis ,Animal Cells ,Immune Physiology ,Medicine and Health Sciences ,Myocyte ,Myocytes, Cardiac ,Flow cytometry ,Biology (General) ,health care economics and organizations ,Cardiomyocytes ,education.field_of_study ,Immune System Proteins ,CD24 ,General Neuroscience ,Cell Differentiation ,Heart ,Cell biology ,Heart / growth & development ,Spectrophotometry ,Myocytes, Cardiac / physiology ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Female ,Cytophotometry ,Anatomy ,Cellular Types ,Single-Cell Analysis ,General Agricultural and Biological Sciences ,Research Article ,CD24 Antigen / physiology ,[SDV.IMM] Life Sciences [q-bio]/Immunology ,Myocardium / metabolism ,QH301-705.5 ,Immunology ,Population ,Cardiology ,Muscle Tissue ,Biology ,Research and Analysis Methods ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,CD24 Antigen / metabolism ,health services administration ,Genetics ,Regeneration ,Animals ,Cell Lineage ,Heart / physiology ,Antigens ,Progenitor cell ,education ,Muscle Cells ,General Immunology and Microbiology ,Cluster of differentiation ,Myocardium ,Biology and Life Sciences ,Proteins ,CD24 Antigen ,Cell adhesion ,Cell Biology ,Molecular Development ,Gene regulation ,Mice, Inbred C57BL ,Transplantation ,Myocardial infarction ,Biological Tissue ,030104 developmental biology ,Myocardial Ischemia / physiopathology ,Regeneration / physiology ,Cardiovascular Anatomy ,Myocardial Ischemia / metabolism ,Adhesion molecules ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
The assessment of the regenerative capacity of the heart has been compromised by the lack of surface signatures to characterize cardiomyocytes (CMs). Here, combined multiparametric surface marker analysis with single-cell transcriptional profiling and in vivo transplantation identify the main mouse fetal cardiac populations and their progenitors (PRGs). We found that CMs at different stages of differentiation coexist during development. We identified a population of immature heat stable antigen (HSA)/ cluster of differentiation 24 (CD24)+ CMs that persists throughout life and that, unlike other CM subsets, actively proliferates up to 1 week of age and engrafts cardiac tissue upon transplantation. In the adult heart, a discrete population of HSA/CD24+ CMs appears as mononucleated cells that increase in frequency after infarction. Our work identified cell surface signatures that allow the prospective isolation of CMs at all developmental stages and the detection of a subset of immature CMs throughout life that, although at reduced frequencies, are poised for activation in response to ischemic stimuli. This work opens new perspectives in the understanding and treatment of heart pathologies., This study identifies HSA/CD24 as a unique marker for immature cardiomyocytes, showing that HSA-positive cardiomyocytes persist throughout life, proliferating within the first week after birth and responding to myocardial infarction in the adult by expanding through cell division.
- Published
- 2019
- Full Text
- View/download PDF
33. Human multilineage pro-epicardium/foregut organoids support the development of an epicardium/myocardium organoid
- Author
-
Mariana A. Branco, Tiago P. Dias, Joaquim M. S. Cabral, Perpetua Pinto-do-Ó, and Maria Margarida Diogo
- Subjects
Science - Abstract
Stem cell models of organogenesis are a valuable tool for the study of human development, but often lack the context of tissue-tissue interaction. Here they generate human multi-lineage organoids comprising pro-epicardium, septum transversum, and liver bud, which they co-culture with heart organoids to generate a physiologically relevant model of organogenesis.
- Published
- 2022
- Full Text
- View/download PDF
34. Three-dimensional scaffolds of fetal decellularized hearts exhibit enhanced potential to support cardiac cells in comparison to the adult
- Author
-
Mário A. Barbosa, Ana Catarina Silva, Diana S. Nascimento, Maria José Oliveira, Sólveig Thorsteinsdóttir, Tatiana P. Resende, Andreia Nunes, Joana Caldeira, Vasco Sampaio-Pinto, Perpétua Pinto-do-Ó, S.C. Rodrigues, Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto = University of Porto, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Instituto de Engenharia Biomédica (INEB), Centre for Ecology, Evolution and Environmental Change [Lisboa], Universidade de Lisboa = University of Lisbon (ULISBOA), Lymphopoïèse (Lymphopoïèse (UMR_1223 / U1223 / U-Pasteur_4)), Institut Pasteur [Paris] (IP)-Université Paris Diderot - Paris 7 (UPD7)-Institut National de la Santé et de la Recherche Médicale (INSERM), This work was financed by PTDC/SAUORG/118297/2010, FEDER - Fundo Europeu de Desenvolvimento Regional funds through the COMPETE 2020 - Operacional Programme for Competitiveness and Internationalisation (POCI), Portugal 2020, and by Portuguese funds through FCT - Fundação para a Ciência e a Tecnologia/Ministério da Ciência, Tecnologia e Inovação in the framework of the project 'Institute for Research and Innovation in Health Sciences'- POCI-01-0145-FEDER-007274 and individual fellowships (SFRH/BD/88780/2012 to A.C.S., BI-PTDC/SAU-ORG/118297/2010 to A.M.N., SFRH/BPD/78187/2011 to J.C., SFRH/BPD/80588/2011 to T.P.R and SFRH/BD/111799/2015 to V.S.P.). PPO was recipient of an invited scientist grant by Institut Pasteur, Paris, France. MJO is Portuguese Science Foundation Fellow (FCT-Investigator 2012)., European Project: 118297,FCT::,PTDC/2010,PTDC/SAU-ORG/118297/2010(2012), European Project: SFRH/BD/88780/2012,FCT::Doutoramento,SFRH/2012,SFRH/BD/88780/2012(2013), European Project: SFRH/BPD/78187/2011,FCT::Pós-Doutoramento,SFRH/2011,SFRH/BPD/78187/2011(2012), European Project: SFRH/BPD/80588/2011,FCT::Pós-Doutoramento,SFRH/2011,SFRH/BPD/80588/2011(2012), Universidade do Porto [Porto], centre for ecology, evolution and environmental change, Universidade de Lisboa, Centro de Biologia Ambiental/Departamento de Biologia Animal, University of Porto, Institut Pasteur [Paris]-Université Paris Diderot - Paris 7 (UPD7)-Institut National de la Santé et de la Recherche Médicale (INSERM), Universidade do Porto, and Universidade de Lisboa (ULISBOA)
- Subjects
0301 basic medicine ,Scaffold ,Aging ,Cell Survival ,3D scaffolds ,[SDV]Life Sciences [q-bio] ,Biophysics ,Bioengineering ,Fetal microenvironments ,030204 cardiovascular system & hematology ,Cardiac tissue engineering ,Biomaterials ,Extracellular matrix ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Fetal Heart ,Tissue engineering ,[SDV.MHEP.CSC]Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,Cell Movement ,Animals ,Myocytes, Cardiac ,Progenitor cell ,Cells, Cultured ,Decellularization ,Three dimensional scaffolds ,Fetus ,Cell-Free System ,Tissue Engineering ,Tissue Scaffolds ,Chemistry ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Mechanics of Materials ,Printing, Three-Dimensional ,Ceramics and Composites ,Feasibility Studies ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Biomedical engineering ,Explant culture - Abstract
International audience; A main challenge in cardiac tissue engineering is the limited data on microenvironmental cues that sustain survival, proliferation and functional proficiency of cardiac cells. The aim of our study was to evaluate the potential of fetal (E18) and adult myocardial extracellular matrix (ECM) to support cardiac cells. Acellular three-dimensional (3D) bioscaffolds were obtained by parallel decellularization of fetal- and adult-heart explants thereby ensuring reliable comparison. Acellular scaffolds retained main constituents of the cardiac ECM including distinctive biochemical and structural meshwork features of the native equivalents. In vitro, fetal and adult ECM-matrices supported 3D culture of heart-derived Sca-1(+) progenitors and of neonatal cardiomyocytes, which migrated toward the center of the scaffold and displayed elongated morphology and excellent viability. At the culture end-point, more Sca-1(+) cells and cardiomyocytes were found adhered and inside fetal bioscaffolds, compared to the adult. Higher repopulation yields of Sca-1(+) cells on fetal ECM relied on β1-integrin independent mitogenic signals. Sca-1(+) cells on fetal bioscaffolds showed a gene expression profile that anticipates the synthesis of a permissive microenvironment for cardiomyogenesis. Our findings demonstrate the superior potential of the 3D fetal microenvironment to support and instruct cardiac cells. This knowledge should be integrated in the design of next-generation biomimetic materials for heart repair.
- Published
- 2016
- Full Text
- View/download PDF
35. Abstract 18331: Endothelial Microrna-155 Promotes Myocardial Microvascular Permeability and Inflammatory Cell Adhesion in Experimental Septic Cardiomyopathy
- Author
-
Adelino F. Leite-Moreira, Sara Ribeiro, Fabiana Baganha, Rui J Cerqueira, Francisco Vasques-Nóvoa, Diana S. Nascimento, Perpétua Pinto-do-Ó, Roberto Roncon-Albuquerque, Catarina Quina-Rodrigues, and Tiago L. Laundos
- Subjects
Cardiac function curve ,CD31 ,Cell adhesion molecule ,business.industry ,medicine.medical_treatment ,Inflammation ,Vascular permeability ,Adhesion ,Pharmacology ,Nitric oxide ,chemistry.chemical_compound ,Cytokine ,chemistry ,Physiology (medical) ,medicine ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business - Abstract
Introduction: Septic cardiomyopathy remains a leading cause of death in critically ill patients. Myocardial edema and inflammatory cell recruitment to cardiac interstitial space are proposed mechanisms underlying sepsis-induced contractile dysfunction. Hypothesis: Evaluate the role of endothelial microRNA-155 (miR-155) in experimental septic cardiomyopathy. Methods: Experimental sepsis was induced using LPS injection in WT (n=20) and miR-155-/- (KO; n=20) male mice. Cardiac function was evaluated through echocardiography. Expression of NOS2, NOS3, VASP phosphorylation and myocardial nitrite/nitrate content was evaluated. Cardiac microvascular (MV) permeability was assessed with evans blue-albumin conjugate leaking to extravascular space and myocardial water content. Mouse cardiac MV endothelial cells (MCMVEC; CD31+Sca-1+CD45-) were isolated with FACS. Quantification of miR-155, cytokine mRNA profile and adhesion molecules (ICAM1, VCAM1 and E-selectin) was performed. Human cardiac MV endothelial cells (HCMVEC) were transfected with anti-miR-155 or scramble LNA probe and incubated with LPS (100ng/mL) or vehicle for 24h. Evaluation of cytokine mRNA profile, NOS isoforms, VASP phosphorylation and adhesion molecules was performed. Monocyte adhesion assay was performed using Dil-stained THP1 monocytic cells. Results: LPS exposure increased miR-155 expression in the myocardium, MCMVEC and HCMVEC. KO mice presented with attenuated LPS-induced cardiac dysfunction, MV permeability, NOS2 expression and VASP phosphorylation, compared with WT. KO MCMVEC presented with attenuated LPS-induced cytokine expression and adhesion molecules, compared with WT cells. Mir-155 inhibition in HCMVEC prevented LPS-induced overexpression of cytokines and adhesion molecules, nitric oxide production and monocyte adhesion, through SOCS1-mediated inhibition of STAT3 and NF-kB signaling pathways. Conclusions: Endothelial miR-155 is upregulated in experimental septic cardiomyopathy. MiR-155 knockout attenuates LPS-induced myocardial dysfunction, edema and MV permeability. In MCMVEC and HCMVEC, miR-155 inhibition blunted LPS-induced nitric oxide production, cytokine and cell adhesion molecules overexpression.
- Published
- 2015
- Full Text
- View/download PDF
36. Three-dimensional spheroid cell culture of umbilical cord tissue-derived mesenchymal stromal cells leads to enhanced paracrine induction of wound healing
- Author
-
Pedro E. Cruz, Jorge M. Santos, Mariana Filipe, Madalena Cipriano, Diogo Mosqueira, Diana S. Nascimento, Elysse C. Filipe, Manuela Gaspar, Sandra Simões, Rita N. Bárcia, Matilde Castro, Mariana Teixeira, S.P. Camões, Joana P. Miranda, Helder Cruz, Perpétua Pinto-do-Ó, ECBio – Investigação e Desenvolvimento em Biotecnologia S.A., Instituto de Medicina Molecular (iMM), Faculdade de Medicina [Lisboa], Universidade de Lisboa (ULISBOA)-Universidade de Lisboa (ULISBOA), Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Instituto de Engenharia Biomédica (INEB), Lymphopoïèse, Institut Pasteur [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Instituto de Ciencias Biomedicas Abel Salazar (ICBAS), This work was supported by FCT-Fundação para a Ciência e a Tecnologia [EXPL/DTP-FTO/0308/2013, SFRH/BPD/96719/2013 and Ciência2008 to JPM, SFRH/BPD/42254/2007 and ON.2(NORTE-07-0124-FEDER-000005) to DSN and SFRH/BD/87508/2012 to MC], by ECBio, and by the project on Biomedical Engineering for Regenerative Therapies and Cancer cofunded by 'ON.2 – O Novo Norte' (Programa Operacional Regional do Norte 2014–2020), by Fundo Europeu de Desenvolvimento Regional-FEDER, Programa Operacional Factores de Competitividade- COMPETE and by QREN., Vougny, Marie-Christine, Universidade de Lisboa = University of Lisbon (ULISBOA)-Universidade de Lisboa = University of Lisbon (ULISBOA), Universidade do Porto = University of Porto, and Institut Pasteur [Paris] (IP)-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
Male ,[SDV.IMM] Life Sciences [q-bio]/Immunology ,Cell Culture Techniques ,Medicine (miscellaneous) ,Enzyme-Linked Immunosorbent Assay ,Biology ,Fibroblast growth factor ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Umbilical Cord ,Extracellular matrix ,03 medical and health sciences ,Paracrine signalling ,0302 clinical medicine ,Cell Movement ,Paracrine Communication ,medicine ,Animals ,Cell Lineage ,Rats, Wistar ,Cells, Cultured ,030304 developmental biology ,Cell Proliferation ,0303 health sciences ,Extracellular Matrix Proteins ,Wound Healing ,Research ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Biology ,3. Good health ,Cell biology ,Rats ,Vascular endothelial growth factor A ,Phenotype ,Microscopy, Fluorescence ,Cell culture ,030220 oncology & carcinogenesis ,Molecular Medicine ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Intercellular Signaling Peptides and Proteins ,Hepatocyte growth factor ,Wound healing ,medicine.drug - Abstract
Introduction The secretion of trophic factors by mesenchymal stromal cells has gained increased interest given the benefits it may bring to the treatment of a variety of traumatic injuries such as skin wounds. Herein, we report on a three-dimensional culture-based method to improve the paracrine activity of a specific population of umbilical cord tissue-derived mesenchymal stromal cells (UCX®) towards the application of conditioned medium for the treatment of cutaneous wounds. Methods A UCX® three-dimensional culture model was developed and characterized with respect to spheroid formation, cell phenotype and cell viability. The secretion by UCX® spheroids of extracellular matrix proteins and trophic factors involved in the wound-healing process was analysed. The skin regenerative potential of UCX® three-dimensional culture-derived conditioned medium (CM3D) was also assessed in vitro and in vivo against UCX® two-dimensional culture-derived conditioned medium (CM2D) using scratch and tubulogenesis assays and a rat wound splinting model, respectively. Results UCX® spheroids kept in our three-dimensional system remained viable and multipotent and secreted considerable amounts of vascular endothelial growth factor A, which was undetected in two-dimensional cultures, and higher amounts of matrix metalloproteinase-2, matrix metalloproteinase-9, hepatocyte growth factor, transforming growth factor β1, granulocyte-colony stimulating factor, fibroblast growth factor 2 and interleukin-6, when compared to CM2D. Furthermore, CM3D significantly enhanced elastin production and migration of keratinocytes and fibroblasts in vitro. In turn, tubulogenesis assays revealed increased capillary maturation in the presence of CM3D, as seen by a significant increase in capillary thickness and length when compared to CM2D, and increased branching points and capillary number when compared to basal medium. Finally, CM3D-treated wounds presented signs of faster and better resolution when compared to untreated and CM2D-treated wounds in vivo. Although CM2D proved to be beneficial, CM3D-treated wounds revealed a completely regenerated tissue by day 14 after excisions, with a more mature vascular system already showing glands and hair follicles. Conclusions This work unravels an important alternative to the use of cells in the final formulation of advanced therapy medicinal products by providing a proof of concept that a reproducible system for the production of UCX®-conditioned medium can be used to prime a secretome for eventual clinical applications. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0082-5) contains supplementary material, which is available to authorized users.
- Published
- 2015
- Full Text
- View/download PDF
37. Hematopoietic progenitor/stem cells immortalized byLhx2 generate functional hematopoietic cells in vivo
- Author
-
Karin Richter, Perpétua Pinto do Ó, and Leif Carlsson
- Subjects
Genotype ,LIM-Homeodomain Proteins ,Immunology ,Biology ,Biochemistry ,CXCR4 ,Colony-Forming Units Assay ,Embryonic and Fetal Development ,Mice ,medicine ,Animals ,Bone Marrow Transplantation ,Cell Line, Transformed ,Interleukin 3 ,Homeodomain Proteins ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,Embryonic stem cell ,Hematopoiesis ,Cell biology ,Mice, Inbred C57BL ,Endothelial stem cell ,Kinetics ,Haematopoiesis ,medicine.anatomical_structure ,Bone marrow ,Stem cell ,Transcription Factors ,Adult stem cell - Abstract
Hematopoietic stem cells (HSCs) are unique in their capacity to maintain blood formation following transplantation into immunocompromised hosts. Expansion of HSCs in vitro is therefore important for many clinical applications but has met with limited success because the mechanisms regulating the self-renewal process are poorly defined. We have previously shown that expression of the LIM-homeobox gene Lhx2 in hematopoietic progenitor cells derived from embryonic stem cells differentiated in vitro generates immortalized multipotent hematopoietic progenitor cell lines. However, HSCs of early embryonic origin, including those derived from differentiated embryonic stem cells, are inefficient in engrafting adult recipients upon transplantation. To address whetherLhx2 can immortalize hematopoietic progenitor/stem cells that can engraft adult recipients, we expressed Lhx2 in hematopoietic progenitor/stem cells derived from adult bone marrow. This approach allowed for the generation of immortalized growth factor–dependent hematopoietic progenitor/stem cell lines that can generate erythroid, myeloid, and lymphoid cells upon transplantation into lethally irradiated mice. When transplanted into stem cell–deficient mice, these cell lines can generate a significant proportion of circulating erythrocytes in primary, secondary, and tertiary recipients for at least 18 months. Thus, Lhx2immortalizes multipotent hematopoietic progenitor/stem cells that can generate functional progeny following transplantation into lethally irradiated hosts and can long-term repopulate stem cell–deficient hosts.
- Published
- 2002
- Full Text
- View/download PDF
38. Sca-1+ Cardiac Progenitor Cells and Heart-Making: A Critical Synopsis
- Author
-
Ana Cumano, Mariana Valente, Perpétua Pinto-do-Ó, Diana S. Nascimento, Stem-Cell Microenvironments in Repair/Regeneration Team, Microenvironments for NewTherapies Group, Universidade do Porto-INEB - Instituto Nacional de Engenharia Biomedica, Lymphopoïèse, Institut Pasteur [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Instituto de Ciencias Biomedicas Abel Salazar (ICBAS), Universidade do Porto, This work was supported by Fundacao para a Ciencia e a Tecnologia [SFRH/BD/74218/2010] to M.V., [SFRH/BPD/42254/2007] and QREN/ON.2 [NORTE-07-0124-FEDER-000005] to D.S.N., and Fundo Europeu de Desenvolvimento Regional, Programa Operacional Factores de Competitividade-COMPETE, Quadro de Referencia Estrategico Nacional, Fundo Social Europeu [PEst-C/SAU/LA0002/2013, PTDC/ SAU-ORG/118297/2010, and NORTE-07-0124-FEDER-000005]. By the Pasteur Institute, INSERM, ANR through a grant 'Lymphopoiesis' and through the REVIVE Future Investment Program, La Ligue contre le Cancer with grants to A.C.., Universidade do Porto = University of Porto-INEB - Instituto Nacional de Engenharia Biomedica, Institut Pasteur [Paris] (IP)-Institut National de la Santé et de la Recherche Médicale (INSERM), Universidade do Porto = University of Porto, and Vougny, Marie-Christine
- Subjects
[SDV.IMM] Life Sciences [q-bio]/Immunology ,Cellular differentiation ,Comprehensive Review ,030204 cardiovascular system & hematology ,Biology ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Myocyte ,Animals ,Antigens, Ly ,Humans ,Myocytes, Cardiac ,Progenitor cell ,030304 developmental biology ,0303 health sciences ,Stem Cells ,Mesenchymal stem cell ,Cell Differentiation ,Heart ,Cell Biology ,Hematology ,Embryonic stem cell ,3. Good health ,Cell biology ,Endothelial stem cell ,Immunology ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Stem cell ,Developmental Biology - Abstract
International audience; The identification, in the adult, of cardiomyocyte turnover events and of cardiac progenitor cells (CPCs) has revolutionized the field of cardiovascular medicine. However, the low rate of CPCs differentiation events reported both in vitro and in vivo, even after injury, raised concerns on the biological significance of these subsets. In this Comprehensive Review, we discuss the current understanding of cardiac Lin(-)Sca-1(+) cells in light of what is also known for cellular compartments with similar phenotypes in other organs. The Lin(-)Sca-1(+) heart subset is heterogeneous and displays a mesenchymal profile, characterized by a limited ability to generate cardiomyocytes in vitro and in vivo, even after injury. There is no evidence for Sca-1 expression in embryonic cardiovascular progenitors. In other organs, Sca-1 expression is mainly observed on mesoderm-derived cells, although it is not restricted to stem/progenitor cell populations. It is urgent to determine, at a single cell level, to which extent cardiac Lin(-)Sca-1(+) cells overlap with the fibroblast compartment.
- Published
- 2014
- Full Text
- View/download PDF
39. Hippo Pathway Effectors Control Cardiac Progenitor Cell Fate by Acting as Dynamic Sensors of Substrate Mechanics and Nanostructure
- Author
-
Jun Nakanishi, Diogo Mosqueira, Enrico Traversa, Perpétua Pinto-do-Ó, Marie-José Goumans, Mitsuhiro Ebara, Stefania Pagliari, Sara Romanazzo, Koichiro Uto, Carmen Escobedo-Lucea, Paolo Di Nardo, Ornella Franzese, Giancarlo Forte, Akiyoshi Taniguchi, and Takao Aoyagi
- Subjects
Settore MED/09 - Medicina Interna ,Intracellular Space ,Myocardial Infarction ,cardiac differentiation ,General Physics and Astronomy ,02 engineering and technology ,substrate nanotopography ,mechano-transduction ,Extracellular matrix ,Cell Movement ,General Materials Science ,0303 health sciences ,Effector ,General Engineering ,Signal transducing adaptor protein ,Adaptor Proteins ,Mechanics ,021001 nanoscience & nanotechnology ,Protein-Serine-Threonine Kinases ,Biomechanical Phenomena ,Extracellular Matrix ,Adult Stem Cells ,Signal transduction ,Stem cell ,0210 nano-technology ,Mechanical Processes ,adult cardiac progenitor cell ,Signal Transduction ,Adult ,Biology ,Protein Serine-Threonine Kinases ,03 medical and health sciences ,Humans ,YAP/TAZ ,Hippo Signaling Pathway ,Progenitor cell ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Mechanical Phenomena ,Hippo signaling pathway ,Cell growth ,Myocardium ,Signal Transducing ,Nanostructures ,Phosphoproteins ,Transcription Factors ,YAP-Signaling Proteins ,Acyltransferases - Abstract
Stem cell responsiveness to extracellular matrix (ECM) composition and mechanical cues has been the subject of a number of investigations so far, yet the molecular mechanisms underlying stem cell mechano-biology still need full clarification. Here we demonstrate that the paralog proteins YAP and TAZ exert a crucial role in adult cardiac progenitor cell mechano-sensing and fate decision. Cardiac progenitors respond to dynamic modifications in substrate rigidity and nanopattern by promptly changing YAP/TAZ intracellular localization. We identify a novel activity of YAP and TAZ in the regulation of tubulogenesis in 3D environments and highlight a role for YAP/TAZ in cardiac progenitor proliferation and differentiation. Furthermore, we show that YAP/TAZ expression is triggered in the heart cells located at the infarct border zone. Our results suggest a fundamental role for the YAP/TAZ axis in the response of resident progenitor cells to the modifications in microenvironment nanostructure and mechanics, thereby contributing to the maintenance of myocardial homeostasis in the adult heart. These proteins are indicated as potential targets to control cardiac progenitor cell fate by materials design.
- Published
- 2014
40. Human umbilical cord tissue-derived mesenchymal stromal cells attenuate remodeling after myocardial infarction by proangiogenic, antiapoptotic, and endogenous cell-activation mechanisms
- Author
-
Tatiana P. Resende, Diogo Mosqueira, Ana Francisca Araújo, Mariana Teixeira, Diana S. Nascimento, José Paulo Martins, Mariana Filipe, Rita N. Bárcia, Mariana Valente, Jorge M. Santos, Pedro E. Cruz, Luís Moura Sousa, Helder Cruz, Perpétua Pinto-do-Ó, and Joana Almeida
- Subjects
Angiogenesis ,Myocardial Infarction ,Neovascularization, Physiologic ,Medicine (miscellaneous) ,Apoptosis ,Biology ,Mesenchymal Stem Cell Transplantation ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Umbilical vein ,Cell Line ,Mice ,Paracrine Communication ,Human Umbilical Vein Endothelial Cells ,medicine ,Animals ,Humans ,Myocytes, Cardiac ,Progenitor cell ,Ventricular remodeling ,Cells, Cultured ,Cell Proliferation ,Ventricular Remodeling ,Research ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Biology ,Fetal Blood ,medicine.disease ,Mice, Inbred C57BL ,Transplantation ,Immunology ,Cancer research ,Molecular Medicine ,Female ,Stem cell ,Cell activation - Abstract
Introduction: Among the plethora of cells under investigation to restore a functional myocardium, mesenchymal stromal cells (MSCs) have been granted considerable interest. However, whereas the beneficial effects of bone marrow MSCs (BM-MSCs) in the context of the diseased heart are widely reported, data are still scarce on MSCs from the umbilical cord matrix (UCM-MSCs). Herein we report on the effect of UCM-MSC transplantation to the infarcted murine heart, seconded by the dissection of the molecular mechanisms at play. Methods: Human umbilical cord tissue-derived MSCs (UCX®), obtained by using a proprietary technology developed by ECBio, were delivered via intramyocardial injection to C57BL/6 females subjected to permanent ligation of the left descending coronary artery. Moreover, medium produced by cultured UCX® preconditioned under normoxia (CM) or hypoxia (CMH) was collected for subsequent in vitro assays. Results: Evaluation of the effects upon intramyocardial transplantation shows that UCX® preserved cardiac function and attenuated cardiac remodeling subsequent to myocardial infarction (MI). UCX® further led to increased capillary density and decreased apoptosis in the injured tissue. In vitro, UCX®-conditioned medium displayed (a) proangiogenic activity by promoting the formation of capillary-like structures by human umbilical vein endothelial cells (HUVECs), and (b) antiapoptotic activity in HL-1 cardiomyocytes subjected to hypoxia. Moreover, in adult murine cardiac Sca-1 + progenitor cells (CPCs), conditioned medium enhanced mitogenic activity while activating a gene program characteristic of cardiomyogenic differentiation. Conclusions: UCX® preserve cardiac function after intramyocardial transplantation in a MI murine model. The cardioprotective effects of UCX® were attributed to paracrine mechanisms that appear to enhance angiogenesis, limit the extent of the apoptosis, augment proliferation, and activate a pool of resident CPCs. Overall, these results suggest that UCX® should be considered an alternative cell source when designing new therapeutic approaches to treat MI.
- Published
- 2014
- Full Text
- View/download PDF
41. Building and Repairing the Heart: What Can We Learn from Embryonic Development?
- Author
-
Perpétua Pinto-do-Ó, Ana G. Freire, and Tatiana P. Resende
- Subjects
Heart Injury ,Organogenesis ,Cell ,Embryonic Development ,lcsh:Medicine ,Disease ,Review Article ,030204 cardiovascular system & hematology ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Stress, Physiological ,medicine ,Animals ,Humans ,030304 developmental biology ,0303 health sciences ,General Immunology and Microbiology ,Heart development ,Regeneration (biology) ,Myocardium ,Embryogenesis ,lcsh:R ,Heart ,General Medicine ,Anatomy ,Embryonic stem cell ,medicine.anatomical_structure ,Signal transduction ,Neuroscience - Abstract
Mammalian heart formation is a complex morphogenetic event that depends on the correct temporal and spatial contribution of distinct cell sources. During cardiac formation, cellular specification, differentiation, and rearrangement are tightly regulated by an intricate signaling network. Over the last years, many aspects of this network have been uncovered not only due to advances in cardiac development comprehension but also due to the use of embryonic stem cells (ESCs)in vitromodel system. Additionally, several of these pathways have been shown to be functional or reactivated in the setting of cardiac disease. Knowledge withdrawn from studying heart development, ESCs differentiation, and cardiac pathophysiology may be helpful to envisage new strategies for improved cardiac repair/regeneration. In this review, we provide a comparative synopsis of the major signaling pathways required for cardiac lineage commitment in the embryo and murine ESCs. The involvement and possible reactivation of these pathways following heart injury and their role in tissue recovery will also be discussed.
- Published
- 2014
42. Multipotent hematopoietic progenitor cells immortalized by Lhx2 self-renew by a cell nonautonomous mechanism
- Author
-
Ewa Wandzioch, Perpétua Pinto do Ó, Leif Carlsson, and Åsa Kolterud
- Subjects
Cancer Research ,Time Factors ,LIM-Homeodomain Proteins ,Cell ,Cell Culture Techniques ,Biology ,Transfection ,Mice ,Colony-Stimulating Factors ,Tumor Cells, Cultured ,Genetics ,medicine ,Animals ,Humans ,Growth Substances ,Molecular Biology ,Cells, Cultured ,Cell Line, Transformed ,Homeodomain Proteins ,Mechanism (biology) ,Interleukins ,Stem Cells ,Hematopoietic Tissue ,hemic and immune systems ,Cell Biology ,Hematology ,Embryo, Mammalian ,Hematopoietic Stem Cells ,Recombinant Proteins ,Cell biology ,Endothelial stem cell ,Kinetics ,Haematopoiesis ,medicine.anatomical_structure ,Culture Media, Conditioned ,Cytokines ,Hemangioblast ,Interleukin-3 ,Stem cell ,Multiple Myeloma ,Cell Division ,Transcription Factors ,Adult stem cell - Abstract
Direct molecular and cellular studies of hematopoietic stem cells (HSCs) are hampered by the low levels of HSCs in hematopoietic tissues. To address these issues, we generated immortalized multipotent hematopoietic precursor cell (HPC) lines by expressing the LIM-homeobox gene Lhx2 (previously LH2) in hematopoietic progenitors derived from embryonic stem cells differentiated in vitro.To validate further the relevance of the HPC lines as a model for normal HSCs, we analyzed in detail the growth requirements of HPC lines in vitro.Lhx2 immortalized the HPC lines by a putatively novel and cell nonautonomous mechanism. Self-renewal of the HPC lines is dependent on functional Lhx2 expression. Most early-acting hematopoiesis-related growth factors show synergistic effects on the HPC lines, whereas late-acting factors do not induce differentiation by themselves. Transforming growth factor-beta(1) is a potent inhibitor of proliferation of the HPC lines. HPC lines form cobblestone areas with high efficiency when seeded onto stromal cell lines, and the cobblestone area-forming cell can be maintained in these cultures for several months.Our data show that, in many respects, HPC lines are similar to normal hematopoietic progenitor/stem cells on the cellular level, in contrast to most previously described multipotent hematopoietic cell lines. The cell nonautonomous mechanism for immortalization of the HPC lines suggests that Lhx2 regulates, directly or indirectly, soluble mediators involved in self-renewal of the HPC lines.
- Published
- 2001
- Full Text
- View/download PDF
43. Expression of the LIM-homeobox gene LH2 generates immortalized Steel factor-dependent multipotent hematopoietic precursors
- Author
-
Åsa Kolterud, Perpétua Pinto do Ó, and Leif Carlsson
- Subjects
Cellular differentiation ,Genetic Vectors ,LIM-Homeodomain Proteins ,Down-Regulation ,Stem cell factor ,Biology ,Cell fate determination ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Transformation, Genetic ,Animals ,Cell Lineage ,RNA, Messenger ,Progenitor cell ,Molecular Biology ,Cell Line, Transformed ,Homeodomain Proteins ,Stem Cell Factor ,General Immunology and Microbiology ,General Neuroscience ,Genes, Homeobox ,Cell Differentiation ,Hematopoietic Stem Cells ,Antigens, Differentiation ,Embryonic stem cell ,Molecular biology ,Recombinant Proteins ,Hematopoiesis ,Cell biology ,Haematopoiesis ,Retroviridae ,Cell culture ,Hemangioblast ,Transcription Factors ,Research Article - Abstract
The genes controlling self-renewal and differentiation in the hematopoietic system are largely unknown. The LIM-homeobox genes are known to be important for asymmetric cell divisions and differentiation of specific cell types and organs. One member of this family, LH2, is expressed in fetal liver at the time of active hematopoiesis. Therefore, we have assessed the function of LH2 during the formation and initial expansion of the hematopoietic system by differentiating LH2-transduced embryonic stem (ES) cells in vitro. This procedure generated multipotent hematopoietic precursor cell (HPC) lines that required Steel factor for growth. HPC lines have been maintained in an undifferentiated state in culture for >7 months. Other growth factors tested efficiently induce terminal differentiation of HPCs into various mature myeloid lineages. Steel factor is also required and acts synergistically with the other growth factors to generate multilineage colonies from the HPCs. These HPC lines express transcription factors that are consistent with an immature progenitor, and the pattern of cell surface marker expression is similar to that of early fetal multipotent hematopoietic progenitors. Collectively, these data suggest that the HPC lines represent an early fetal multipotent hematopoietic progenitor, and suggest a role for LH2 in the control of cell fate decision and/or proliferation in the hematopoietic system.
- Published
- 1998
- Full Text
- View/download PDF
44. Antagonistic actions of renal dopamine and 5-hydroxytryptamine: effects of amine precursors on the cell inward transfer and decarboxylation
- Author
-
Perpétua Pinto-do-Ó and Patrício Soares-da-Silva
- Subjects
Male ,Serotonin ,Carboxy-lyases ,Stereochemistry ,Decarboxylation ,In Vitro Techniques ,Renal Agents ,Medicinal chemistry ,5-Hydroxytryptophan ,Kidney Tubules, Proximal ,Levodopa ,chemistry.chemical_compound ,medicine ,Animals ,Rats, Wistar ,Pharmacology ,Aromatic L-amino acid decarboxylase ,Kidney ,Dose-Response Relationship, Drug ,biology ,Probenecid ,Decynium-22 ,Rats ,Dose–response relationship ,medicine.anatomical_structure ,chemistry ,Aromatic-L-Amino-Acid Decarboxylases ,biology.protein ,Saturation vapor curve ,Research Article ,Organic anion - Abstract
1. The present work was designed to examine the interference of L-3,4-dihydroxyphenylalanine (L-DOPA) on the cell inward transport of L-5-hydroxytryptophan (L-5-HTP) and on its decarboxylation by aromatic L-amino acid decarboxylase (AAAD) in rat isolated renal tubules. 2. The accumulation of both L-5-HTP and L-DOPA in renal tubules was found to occur through non-saturable and saturable mechanisms. The kinetics of the saturable component L-5-HTP and L-DOPA uptake in renal tubules were as follows: L-5-HTP, Vmax = 24.9 +/- 4.5 nmol mg-1 protein h-1 and Km = 121 (95% confidence limits: 75, 193) microM (n = 5); L-DOPA, Vmax = 58.0 +/- 4.3 nmol mg-1 protein h-1 and Km = 135 (97, 188) microM (n = 5). When the saturation curve of L-5-HTP tubular uptake was performed in the presence of L-DOPA (250 microM), the maximal rate of accumulation of L-5-HTP in renal tubules was found to be markedly (P < 0.01) reduced (Vmax = 10.5 +/- 1.7 nmol mg-1 protein h-1, n = 4); this was accompanied by a significant (P < 0.05) increase in Km values (325 [199, 531] microM, n = 4). 3. L-DOPA (50 to 2000 microM) was found to produce a concentration-dependent decrease (38% to 91% reduction) in the tubular uptake of 5-HTP; the Ki value (in microM) of L-DOPA for inhibition of L-5-HTP uptake was found to be 29.1 (13.8, 61.5) (n = 6). 4. At the highest concentration tested the organic anion inhibitor, probenecid (10 microM) produced no significant (P = 0.09) changes in L-5-HTP and L-DOPA uptake (18% and 22% reduction, respectively). The organic cation inhibitor, cyanine 863 (1-ethyl-2-[1,4-dimethyl-2-phenyl-6-pyrimidinylidene)methyl]-quino linium) produced a potent inhibitory effect on the tubular uptake of L-5-HTP (Ki = 212 [35, 1289] nM, n = 8), being slightly less effective against L-DOPA uptake (Ki = 903 [584, 1396] nM, n = 5). The cyanine derivatives 1,1-diethyl-2,4-cyanine (decynium 24) and 1,1-diethyl-2,2-cyanine (decynium 22) potently inhibited the tubular uptake of both L-5-HTP (Ki = 100 [49, 204] and 120 [26, 561] nM, n = 4-6, respectively) and L-DOPA (Ki = 100 [40, 290] and 415 [157, 1094] nM, n = 5, respectively). 5. The Vmax and Km values for AAAD using L-DOPA as the substrate (Vmax = 479.9 +/- 74.0 nmol mg-1 protein h-1; Km = 2380 [1630, 3476] microM; n = 4) were both found to be significantly (P < 0.01) higher than those observed when using L-5-HTP (Vmax = 81.4 +/- 5.2 nmol mg-1 protein h-1, Km = 97 [87, 107] microM, n = 10). The addition of 5 mM L-DOPA to the incubation medium reduced by 30% (P < 0.02) the maximal rate of decarboxylation of L-5-HTP (Vmax = 56.7 +/- 3.1 nmol mg-1 protein h-1, n = 10) and resulted in a significant (P < 0.05) increase in Km values (249 [228, 270] microM, n = 10). 6. The results presented suggest that L-5-HTP and L-DOPA are using the same transporter (most probably, the organic cation transporter) in order to be taken up into renal tubular cells; L-DOPA exerts a competitive type of inhibition upon the tubular uptake and decarboxylation of L-5-HTP. The decrease in the formation of 5-HT as induced by L-DOPA may also depend on a decrease in the rate of its decarboxylation by AAAD.
- Published
- 1996
- Full Text
- View/download PDF
45. Antagonistic actions of renal dopamine and 5- hydroxytryptamine: increase in Na+, K+-ATPase activity in renal proximal tubules via activation of 5-HT1A receptors
- Author
-
Patrício Soares-da-Silva, Perpétua Pinto-do-Ó, and Alejandro M. Bertorello
- Subjects
Male ,Agonist ,Serotonin ,medicine.medical_specialty ,medicine.drug_class ,Dopamine ,Sodium ,chemistry.chemical_element ,Stimulation ,In Vitro Techniques ,Piperazines ,Kidney Tubules, Proximal ,Rats, Sprague-Dawley ,Internal medicine ,medicine ,Animals ,Na+/K+-ATPase ,Receptor ,Pharmacology ,8-Hydroxy-2-(di-n-propylamino)tetralin ,Kidney ,Dose-Response Relationship, Drug ,Receptor antagonist ,Rats ,Serotonin Receptor Agonists ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Receptors, Serotonin ,5-HT1A receptor ,Serotonin Antagonists ,Sodium-Potassium-Exchanging ATPase ,Research Article - Abstract
1. 5-Hydroxytryptamine (5-HT) is antinatriuretic. Since this effect of 5-HT is not accomplished by changes in glomerular haemodynamics, we have examined in this study whether 5-HT may influence sodium excretion by affecting the Na+, K(+)-ATPase activity in renal cortical tubules. 2. Na+, K(+)-ATPase activity was determined as the rate of [32P]-ATP hydrolysis in renal cortical tubules in suspension. Basal Na+, K(+)-ATPase activity in renal tubules was 4.8 +/- 0.4 mumol Pi mg-1 protein h-1 (n = 8). The 5-HT1A receptor agonist, (+/-)-8-hydroxy-2-(di-n-propylamino) tetraline (8-OH-DPAT) (10 to 3000 nM) induced a concentration-dependent increase (P < 0.05) in Na+, K(+)-ATPase activity with an EC50 value of 355 nM (95% confidence limits: 178, 708). Maximal stimulation elicited by 3000 nM of 8-OH-DPAT was antagonized by the selective 5-HT1A receptor antagonist, (+)-WAY 100135 10 to 1000 nM) with an IC50 value of 20 nM (14, 29); 0.3 microM (+)-WAY 100135 completely abolished (P < 0.01) the stimulatory effect of 8-OH-DPAT. The stimulatory effect of 8-OH-DPAT was found to be time-dependent (15 +/- 2% and 66 +/- 7% increase at 2.5 and 5.0 min, respectively). The 5-HT2 receptor agonist alpha-methyl-5-HT (100 to 3000 nM) did not induce any significant changes in Na+, K(+)-ATPase activity (5.0 +/- 1.5 mumol Pi mg-1 protein h-1; n = 4). 3. The stimulatory effect 8-OH-DPAT was absent when homogenates were used. Stimulation occurred at a Vmax concentration (70 mM) of sodium supporting the notion that stimulation occurs independently of increasing sodium permeability. 4. The inhibitory effect of dopamine (P < 0.05) on Na+, K(+)-ATPase activity was blunted by co-incubation with 8-OH-DPAT (0.5 microM). 5. It is concluded that activation of 5-HT1A receptors increases Na+, K(+)-ATPase activity in renal cortical tubules; this effect may represent an important cellular mechanism, at the tubule level, responsible for the antinatriuretic effect of 5-HT.
- Published
- 1996
- Full Text
- View/download PDF
46. Systemic delivery of bone marrow-derived mesenchymal stromal cells diminishes neuropathology in a mouse model of Krabbe's disease
- Author
-
Márcia A. Liz, Vera Sousa, Carla Teixeira, Filipa Franquinho, Mónica Mendes Sousa, Catarina Oliveira Miranda, Perpétua Pinto-do-Ó, Giancarlo Forte, and Paolo Di Nardo
- Subjects
Blotting, Western ,Carbazoles ,Schwann cell ,Bone Marrow Cells ,Mesenchymal Stem Cell Transplantation ,Cell Line ,Indole Alkaloids ,Mice ,Osteogenesis ,medicine ,Animals ,Enzyme Inhibitors ,Krabbe's disease ,Adipogenesis ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Mesenchymal stem cell ,Leukodystrophy ,Mesenchymal Stem Cells ,Cell Biology ,medicine.disease ,Flow Cytometry ,Immunohistochemistry ,Sciatic Nerve ,Cell biology ,Leukodystrophy, Globoid Cell ,Transplantation ,medicine.anatomical_structure ,nervous system ,Peripheral nervous system ,Trk receptor ,Immunology ,biology.protein ,Molecular Medicine ,Developmental Biology ,Neurotrophin - Abstract
In Krabbe's disease, a demyelinating disorder, add-on strategies targeting the peripheral nervous system (PNS) are needed, as it is not corrected by bone-marrow (BM) transplantation. To circumvent this limitation of BM transplantation, we assessed whether i.v. delivery of immortalized EGFP+ BM-derived murine mesenchymal stromal cells (BM-MSCTERT-EGFP) targets the PNS of a Krabbe's disease model, the Twitcher mouse. In vitro, BM-MSCTERT-EGFP retained the phenotype of primary BM-MSC and did not originate tumors upon transplantation in nude mice. In vivo, undifferentiated EGFP+ cells grafted the Twitcher sciatic nerve where an increase in Schwann cell precursors and axonal number was detected. The same effect was observed on BM-MSCTERT-EGFP i.v. delivery following sciatic nerve crush, a model of axonal regeneration. Reiterating the in vivo findings, in a coculture system, BM-MSCTERT-EGFP induced the proliferation of Twitcher-derived Schwann cells and the neurite outgrowth of both Twitcher-derived neurons and wild-type neurons grown in the presence of psychosine, the toxic substrate that accumulates in Krabbe's disease. In vitro, this neuritogenic effect was blocked by K252a, an antagonist of Trk receptors, and by antibody blockage of brain derived neurotrophic factor, a neurotrophin secreted by BM-MSCTERT-EGFP and induced in neighboring Schwann cells. In vivo, BM-MSCTERT-EGFP surmounted the effect of K252a, indicating their ability to act through a neurotrophin-independent mechanism. In summary, i.v. delivery of BM-MSCTERT-EGFP exerts a multilevel effect targeting neurons and Schwann cells, coordinately diminishing neuropathology. Therefore, to specifically target the PNS, MSC should be considered an add-on option to BM transplantation in Krabbe's disease and in other disorders where peripheral axonal loss occurs.
- Published
- 2011
47. Bone Marrow-Derived Endothelial Progenitors Expressing Delta-Like 4 (Dll4) Regulate Tumor Angiogenesis
- Author
-
Leonor Remédio, Francisco Caiado, Hideo Yagita, Sergio Dias, Carla Real, Perpétua Pinto-do-Ó, Antonio Duarte, Cátia Igreja, Cristina Borges, Alexandre Trindade, and NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM)
- Subjects
Angiogenesis ,lcsh:Medicine ,MOUSE EMBRYOS ,UP-REGULATION ,THERAPY ,chemistry.chemical_compound ,Mice ,RENAL-CELL CARCINOMA ,Neoplasms ,Basic Cancer Research ,VASCULATURE ,lcsh:Science ,Multidisciplinary ,biology ,Neovascularization, Pathologic ,Stem Cells ,Intracellular Signaling Peptides and Proteins ,VEGF ,Vascular endothelial growth factor ,Oncology ,cardiovascular system ,Medicine ,GROWTH ,Research Article ,Notch signaling pathway ,INHIBITION ,Bone Marrow Cells ,Vascular endothelial growth inhibitor ,BREAST-CANCER MODEL ,Cell Line, Tumor ,Thrombospondin 1 ,Animals ,Humans ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Tumor hypoxia ,lcsh:R ,Calcium-Binding Proteins ,Endothelial Cells ,Membrane Proteins ,Fibronectin ,Transplantation ,chemistry ,Gene Expression Regulation ,HEMATOPOIETIC STEM ,biology.protein ,Cancer research ,Blood Vessels ,lcsh:Q ,Stem Cell Transplantation - Abstract
Neo-blood vessel growth (angiogenesis), which may involve the activation of pre-existing endothelial cells (EC) and/or the recruitment of bone marrow-derived vascular precursor cells (BM-VPC), is essential for tumor growth. Molecularly, besides the well established roles for Vascular endothelial growth factor (VEGF), recent findings show the Notch signalling pathway, in particular the ligand Delta-like 4 (Dll4), is also essential for adequate tumor angiogenesis; Dll4 inhibition results in impaired, non-functional, angiogenesis and reduced tumor growth. However, the role of BM-VPC in the setting of Notch pathway modulation was not addressed and is the subject of the present report. Here we show that SDF-1 and VEGF, which are produced by tumors, increase Dll4 expression on recruited BM-VPC. Mechanistically, BM-VPC activated, in a Dll4-dependent manner, a transcriptional program on mature EC suggestive of EC activation and stabilization. BM-VPC induced ICAM-2 and Fibronectin expression on EC, an effect that was blocked by a Dll4-specific neutralizing antibody. In vivo, transplantation of BM-VPC with decreased Dll4 into tumor-bearing mice resulted in the formation of microvessels with decreased pericyte coverage and reduced fibronectin expression. Consequently, transplantation of BM-VPC with decreased Dll4 resulted in impaired tumor angiogenesis, increased tumor hypoxia and apoptosis, and decreased tumor growth. Taken together, our data suggests that Dll4 expression by BM-VPC affects their communication with tumor vessel endothelial cells, thereby modulating tumor angiogenesis by affecting vascular stability.
- Published
- 2011
48. MIQuant--semi-automation of infarct size assessment in models of cardiac ischemic injury
- Author
-
Joana G. Guedes, Tiago Esteves, Diana S. Nascimento, Perpétua Pinto-do-Ó, Maria de Fátima de Pina, Pedro Quelhas, Ana G. Freire, and Mariana Valente
- Subjects
Male ,Anatomy and Physiology ,Mouse ,Myocardial Infarction ,Infarction ,lcsh:Medicine ,030204 cardiovascular system & hematology ,Cardiovascular ,Cardiovascular System ,Mice ,0302 clinical medicine ,Morphogenesis ,Myocardial infarction ,lcsh:Science ,0303 health sciences ,Multidisciplinary ,Stem Cells ,Animal Models ,medicine.anatomical_structure ,Cardiology ,Heart Development ,Medicine ,Female ,Research Article ,medicine.medical_specialty ,Histology ,Ischemia ,03 medical and health sciences ,Model Organisms ,Text mining ,Internal medicine ,medicine ,Animals ,Regeneration ,Biology ,030304 developmental biology ,Reproducibility ,business.industry ,lcsh:R ,Computational Biology ,Gold standard (test) ,medicine.disease ,Infarct size ,Surgery ,Mice, Inbred C57BL ,Coronary arteries ,lcsh:Q ,business ,Organism Development ,Software ,Developmental Biology - Abstract
Background The cardiac regenerative potential of newly developed therapies is traditionally evaluated in rodent models of surgically induced myocardial ischemia. A generally accepted key parameter for determining the success of the applied therapy is the infarct size. Although regarded as a gold standard method for infarct size estimation in heart ischemia, histological planimetry is time-consuming and highly variable amongst studies. The purpose of this work is to contribute towards the standardization and simplification of infarct size assessment by providing free access to a novel semi-automated software tool. The acronym MIQuant was attributed to this application. Methodology/Principal Findings Mice were subject to permanent coronary artery ligation and the size of chronic infarcts was estimated by area and midline-length methods using manual planimetry and with MIQuant. Repeatability and reproducibility of MIQuant scores were verified. The validation showed high correlation (rmidline length = 0.981; rarea = 0.970 ) and agreement (Bland-Altman analysis), free from bias for midline length and negligible bias of 1.21% to 3.72% for area quantification. Further analysis demonstrated that MIQuant reduced by 4.5-fold the time spent on the analysis and, importantly, MIQuant effectiveness is independent of user proficiency. The results indicate that MIQuant can be regarded as a better alternative to manual measurement. Conclusions We conclude that MIQuant is a reliable and an easy-to-use software for infarct size quantification. The widespread use of MIQuant will contribute towards the standardization of infarct size assessment across studies and, therefore, to the systematization of the evaluation of cardiac regenerative potential of emerging therapies.
- Published
- 2011
49. TNF-alpha regulates the effects of irradiation in the mouse bone marrow microenvironment
- Author
-
Tânia Carvalho, Ana C. Santos, Catarina Osorio, Cátia Igreja, Sofia Correia, Manuela Ferreira, Ana Sofia Cachaço, Jacinta Serpa, Sergio Dias, Rita Fragoso, Perpétua Pinto-do-Ó, and NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM)
- Subjects
Male ,Vascular Endothelial Growth Factor A ,Angiogenesis ,medicine.medical_treatment ,Blotting, Western ,lcsh:Medicine ,Apoptosis ,Bone Marrow Cells ,Biology ,Cell Line ,Mice ,SDG 3 - Good Health and Well-being ,Bone Marrow ,Precursor cell ,hemic and lymphatic diseases ,medicine ,Leukocytes ,Animals ,Oncology/Myelodysplastic Syndrome and Bone Marrow Diseases ,lcsh:Science ,Oncology/Hematological Malignancies ,Mice, Knockout ,Chemotherapy ,Multidisciplinary ,Neovascularization, Pathologic ,Reverse Transcriptase Polymerase Chain Reaction ,Tumor Necrosis Factor-alpha ,Myelodysplastic syndromes ,lcsh:R ,Cancer ,Hematology/Myelodysplastic Syndrome and Bone Marrow Failure ,medicine.disease ,Flow Cytometry ,Antibodies, Neutralizing ,Matrix Metalloproteinases ,Blood Cell Count ,Enzyme Activation ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Immunology ,lcsh:Q ,Tumor necrosis factor alpha ,Bone marrow ,Megakaryocytes ,Research Article - Abstract
BACKGROUND Secondary bone marrow (BM) myelodysplastic syndromes (MDS) are increasingly common, as a result of radio or chemotherapy administered to a majority of cancer patients. Patients with secondary MDS have increased BM cell apoptosis, which results in BM dysfunction (cytopenias), and an increased risk of developing fatal acute leukemias. In the present study we asked whether TNF-alpha, known to regulate cell apoptosis, could modulate the onset of secondary MDS. PRINCIPAL FINDINGS We show that TNF-alpha is induced by irradiation and regulates BM cells apoptosis in vitro and in vivo. In contrast to irradiated wild type (WT) mice, TNF-alpha deficient (TNF-alpha KO) mice or WT mice treated with a TNF-alpha-neutralizing antibody were partially protected from the apoptotic effects of irradiation. Next we established a 3-cycle irradiation protocol, in which mice were sub-lethally irradiated once monthly over a 3 month period. In this model, irradiated WT mice presented loss of microsatellite markers on BM cells, low white blood cell (WBC) counts, reduced megakaryocyte (MK) and platelet levels (thrombocytopenia) and macrocytic anemia, phenoypes that suggest the irradiation protocol resulted in BM dysfunction with clinical features of MDS. In contrast, TNF-alpha KO mice were protected from the irradiation effects: BM cell apoptosis following irradiation was significantly reduced, concomitant with sustained BM MK numbers and absence of other cytopenias. Moreover, irradiated WT mice with long term (> or = 5 months) BM dysfunction had increased BM angiogenesis, MMPs and VEGF and NFkB p65, suggestive of disease progression. CONCLUSION Taken together, our data shows that TNF-alpha induction following irradiation modulates BM cell apoptosis and is a crucial event in BM dysfunction, secondary MDS onset and progression. publishersversion published
- Published
- 2010
50. The thymus as a target for mycobacterial infections
- Author
-
Pere-Joan Cardona, Perpétua Pinto do Ó, Margarida Correia-Neves, Claudia Nobrega, Rui Appelberg, Susana Roque, Instituto de Ciências Biomédicas Abel Salazar, and Universidade do Minho
- Subjects
Tuberculosis ,T-Lymphocytes ,Immunology ,Medicina Básica [Ciências Médicas] ,Colony Count, Microbial ,Spleen ,Thymus Gland ,Microbiology ,Mycobacterium tuberculosis ,03 medical and health sciences ,Mice ,Imunologia, Medicina básica ,0302 clinical medicine ,Immune system ,Antigen ,thymus ,Immunology, Basic medicine ,Basic medicine [Medical and Health sciences] ,medicine ,Animals ,BCG ,030304 developmental biology ,0303 health sciences ,Mycobacterium bovis ,Mice, Inbred BALB C ,Mycobacterium Infections ,Science & Technology ,biology ,Medicina básica [Ciências médicas e da saúde] ,T lymphocyte ,biology.organism_classification ,medicine.disease ,3. Good health ,Mice, Inbred C57BL ,Infectious Diseases ,medicine.anatomical_structure ,Mice, Inbred DBA ,Organ Specificity ,Ciências Médicas::Medicina Básica ,Female ,030215 immunology ,Mycobacterium ,Mycobacterium avium - Abstract
Mycobacterial infections are among the major health threats worldwide. Ability to fight these infections depends on the host's immune response, particularly on macrophages and T lymphocytes produced by the thymus. Using the mouse as a model, and two different routes of infection (aerogenic or intravenous), we show that the thymus is consistently colonized by Mycobacterium tuberculosis, Mycobacterium avium or Mycobacterium bovis BCG. When compared to organs such as the liver and spleen, the bacterial load reaches a plateau at later time-points after infection. Moreover, in contrast with organs such as the spleen and the lung no granuloma were found in the thymus of mice infected with M. tuberculosis or M. avium. Since T cell differentiation depends, to a large extent, on the antigens encountered within the thymus, infection of this organ might alter the host's immune response to infection. Therefore, from now on, the thymus should be considered in studies addressing the immune response to mycobacterial infection., Portuguese “Fundação para a Ciência e Tecnologia” (FCT) and FEDER (POCTI/MGI/39791/2001) and from The American-Portuguese Biomedical Research Fund, Inc. C. Nobrega and P. Pinto do Ó received fellowships from FCT.
- Published
- 2007
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.