Nuova ricerca

ANNA VALERIA SAMARELLI

Assegnista di ricerca
Dipartimento di Scienze Mediche e Chirurgiche Materno-Infantili e dell'Adulto


Home |


Pubblicazioni

2024 - Proteomics Analysis of Formalin-Fixed Paraffine-Embedded Tissue Reveals Key Proteins Related to Lung Dysfunction in Idiopathic Pulmonary Fibrosis. [Articolo su rivista]
Samarelli, ANNA VALERIA; Tonelli, Roberto; Raineri, Giulia; Bruzzi, Giulia; Andrisani, Dario; Gozzi, Filippo; Marchioni, Alessandro; Costantini, Matteo; Fabbiani, Luca; Genovese, Filippo; Pinetti, Diego; Manicardi, Linda; Castaniere, Ivana; Masciale, Valentina; Aramini, Beatrice; Tabbi', Luca; Rizzato, Simone; Bettelli, Stefania; Manfredini, Samantha; Dominici, Massimo; Clini, Enrico; Cerri, Stefania
abstract

Idiopathic pulmonary fibrosis (IPF) severely affects the lung leading to aberrant deposition of extracellular matrix and parenchymal stiffness with progressive functional derangement. The limited availability of fresh tissues represents one of the major limitations to study the molecular profiling of IPF lung tissue. The primary aim of this study was to explore the proteomic profiling yield of archived formalin-fixed paraffin-embedded (FFPE) specimens of IPF lung tissues. We further determined the protein expression according to respiratory functional decline at the time of biopsy. The total proteins isolated from 11 FFPE samples of IPF patients compared to 3 FFPE samples from a non-fibrotic lung defined as controls, were subjected to label-free quantitative proteomic analysis by liquid chromatography-mass spectrometry (LC-MS/MS) and resulted in the detection of about 400 proteins. After the pairwise comparison between controls and IPF, functional enrichment analysis identified differentially expressed proteins that were involved in extracellular matrix signaling pathways, focal adhesion and transforming growth factor β (TGF‐β) signaling pathways strongly associated with IPF onset and progression. Five proteins were significantly over-expressed in the lung of IPF patients with either advanced disease stage (Stage II) or impaired pulmonary function (FVC<75, DLco<55) compared to controls; these were lymphocyte cytosolic protein 1 (LCP1), peroxiredoxin-2 (PRDX2), transgelin 2 (TAGLN2), lumican (LUM) and mimecan (OGN) that might play a key role in the fibrogenic processes. Our work showed that the analysis of FFPE samples was able to identify key proteins that might be crucial for the IPF pathogenesis. These proteins are correlated with lung carcinogenesis or involved in the immune landscape of lung cancer, thus making possible common mechanisms between lung carcinogenesis and fibrosis progression, two pathological conditions at risk for each other in the real life.


2023 - Molecular biology and therapeutic targets of primitive tracheal tumors: focus on tumors derived by salivary glands and squamous cell carcinoma. [Articolo su rivista]
Marchioni, Alessandro; Tonelli, Roberto; Samarelli, ANNA VALERIA; Cappiello, Gaia; Andreani, Alessandro; Tabbì, Luca; Livrieri, Francesco; Bosi, Annamaria; Nori, Ottavia; Mattioli, Francesco; Bruzzi, Giulia; Marchioni, Daniele; Clini, Enrico
abstract


2023 - Physiological effects of lung protective ventilation in patients with lung fibrosis and usual interstitial pneumonia pattern versus primary ARDS: a matched-control study. [Articolo su rivista]
Tonelli, Roberto; Grasso, Salvatore; Cortegiani, Andrea; Ball, Lorenzo; Castaniere, Ivana; Tabbì, Luca; Fantini, Riccardo; Andrisani, Dario; Gozzi, Filippo; Moretti, Antonio; Bruzzi, Giulia; Manicardi, Linda; Cerri, Stefania; Samarelli, ANNA VALERIA; Raineri, Giulia; Murgolo, Francesco; Carzoli, Andrea; Di Mussi, Rossella; Busani, Stefano; Rizzoni, Raffaella; Grasselli, Giacomo; Clini, Enrico; Marchioni, Alessandro
abstract

Background- Although patients with interstitial pneumonia pattern (ILD-UIP) and acute exacerbation (AE) leading to severe acute respiratory failure may require invasive mechanical ventilation (MV), physiological data on lung mechanics during MV are lacking. We aimed at describing the physiological effect of lung protective ventilation in patients with AE-ILD-UIP compared with primary ARDS. Methods- Partitioned lung and chest wall mechanics were assessed in a series of AE-ILD-UIP patients matched 1:1 with primary ARDS as controls (based on BMI and PaO2/FiO2 ratio). Three PEEP levels (zero=ZEEP, 4-8 cmH2O=PEEPLOW, and titrated to achieve positive end-expiratory transpulmonary pressure-PL,EE=PEEPTITRATED) were used for measurements. Results- Ten AE-ILD-UIP patients and 10 matched ARDS were included. In AE-ILD-UIP median PL,EE at ZEEP was - 4.3 [-7.6 – -2.3] cmH2O and lung elastance (EL) 44 [40 – 51] cmH2O/L. At PEEPLOW, PL,EE remained negative and EL did not change (p=0.995) versus ZEEP. At PEEPTITRATED, PL,EE increased to 0.8 [0.3 – 1.5] cmH2O and EL to 49 [43 – 59] (p=0.004 and p<0.001 compared to ZEEP and PEEPLOW, respectively). PL decreased at PEEPLOW (p=0.018) and increased at PEEPTITRATED (p=0.003). In matched ARDS control PEEP titration to obtain a positive PL,EE did not result in significant changes in EL and PL. Conclusions- In mechanically ventilated AE-ILD-UIP patients, differently than in patients with primary ARDS, PEEP titrated to obtain a positive PL,EE significantly worsened lung mechanics.


2022 - Biological effects of COVID-19 on lung cancer: can we drive our decisions? [Articolo su rivista]
Aramini, Beatrice; Masciale, Valentina; Samarelli, Anna V.; Tonelli, Roberto; Cerri, Stefania; Clini, Enrico; Stella, Franco; Dominici, Massimo
abstract

COVID-19 infection caused by SARS-CoV-2 is considered catastrophic because it affects multiple organs, particularly those of the respiratory tract. Although the consequences of this infection are not fully clear, it causes damage to the lungs, the cardiovascular and nervous systems, and other organs, subsequently inducing organ failure. In particular, the effects of SARS-CoV-2-induced inflammation on cancer cells and the tumor microenvironment need to be investigated. COVID-19 may alter the tumor microenvironment, promoting cancer cell proliferation and dormant cancer cell (DCC) reawakening. DCCs reawakened upon infection with SARS-CoV-2 can populate the premetastatic niche in the lungs and other organs, leading to tumor dissemination. DCC reawakening and consequent neutrophil and monocyte/macrophage activation with an uncontrolled cascade of pro-inflammatory cytokines are the most severe clinical effects of COVID-19. Moreover, neutrophil extracellular traps have been demonstrated to activate the dissemination of premetastatic cells into the lungs. Further studies are warranted to better define the roles of COVID-19 in inflammation as well as in tumor development and tumor cell metastasis; the results of these studies will aid in the development of further targeted therapies, both for cancer prevention and the treatment of patients with COVID-19.


2022 - Inspiratory effort and respiratory mechanics in spontaneously breathing patients with acute exacerbation of idiopathic pulmonary fibrosis: a retrospective matched control study. [Articolo su rivista]
Tonelli, Roberto; Castaniere, Ivana; Cortegiani, Andrea; Tabbì, Luca; Fantini, Riccardo; Andrisani, Dario; Gozzi, Filippo; Moretti, Antonio; Bruzzi, Giulia; Manicardi, Linda; Cerbone, Caterina; Nani, Chiara; Biagioni, Emanuela; Cerri, Stefania; Samarelli, Valeria; Busani, Stefano; Girardis, Massimo; Marchioni, Alessandro; Clini, Enrico
abstract

Background- Patients with acute exacerbation of idiopathic pulmonary fibrosis (AE-IPF) may experience severe acute respiratory failure, even requiring ventilatory assistance. Physiological data on lung mechanics during these events are lacking. Methods- Patients with AE-IPF admitted to Respiratory Intensive Care Unit to receive noninvasive ventilation (NIV) were retrospectively analyzed. Esophageal pressure swing (ΔP es ) and respiratory mechanics before and 2 hours after NIV start were collected as primary outcome. Correlation between positive end-expiratory pressure (PEEP) levels and changes of dynamic compliance (dynC RS ) and PaO 2 /FiO 2 ratio was assessed. Further, an exploratory comparison with a historic cohort of ARDS patients matched 1:1 by age, sequential organ failure assessment score, body mass index and PaO 2 /FiO 2 level was performed. Results- At baseline, AE-IPF presented high respiratory drive activation with ΔPes = 27 (21–34) cmH2O, respiratory rate (RR) = 34 (30–39) bpm and minute ventilation (VE) = 21 (20–26) L/min. Two hours after NIV application, ΔPes, RR and VE showed a significant reduction (16 [14–24] cmH2O, p<0.0001, 27 [25–30] bpm, p=0.001, and 18 [17–20] L/min, p=0.003, respectively) while no significant change was found for dynamic transpulmonary pressure (27 [21–34] VS 27 [25–36] cmH2O, p=0.2) expiratory tidal volume (Vte) (9.1 [8.7–10.1] VS 9.3 [8.7 – 9.9] mL/kg of predicted boy weight, p=0.2), dynCRS (28 [19–31] VS 26 [18–28] mL/cmH2O, p=0.1) and dynamic mechanical power (71 [49–94] VS 60 [51–74] J/min, p=0.1). PEEP levels negatively correlated with PaO 2 /FiO 2 ratio and dynC RS (r=–0.67, p=0.03 and r=–0.27, p=0.4, respectively). When compared to AE-IPF, ARDS patients presented lower baseline ΔP es , RR, VE and dynamic mechanical power. At difference with AE-IPF, Vte and dynC RS increased significantly following NIV (p=0.01 and p=0.004 respectively) with PEEP levels directly associated with PaO 2 /FiO 2 ratio and dynC RS (r=0.24, p=0.5 and r=0.65, p=0.04, respectively). Conclusions- In this study, patients with AE-IPF showed a high inspiratory effort, whose intensity was reduced by NIV application without significant improvement in respiratory mechanics. In an exploratory analysis, AE-IPF patients showed a different mechanical behavior under spontaneous unassisted and assisted breathing compared with ARDS of similar severity.


2022 - Molecular Mechanisms and Physiological Changes behind Benign Tracheal and Subglottic Stenosis in Adults [Articolo su rivista]
Marchioni, Alessandro; Tonelli, Roberto; Andreani, Alessandro; Cappiello, Gaia Francesca; Fermi, Matteo; Trentacosti, Fabiana; Castaniere, Ivana; Fantini, Riccardo; Tabbì, Luca; Andrisani, Dario; Gozzi, Filippo; Bruzzi, Giulia; Manicardi, Linda; Moretti, Antonio; Baroncini, Serena; Samarelli, ANNA VALERIA; Marchioni, Daniele; Pinelli, Massimo; DE SANTIS, Giorgio; Stefani, Alessandro; Mattioli, Francesco; Clini, Enrico
abstract

Laryngotracheal stenosis (LTS) is a complex and heterogeneous disease whose pathogenesis remains unclear. LTS is considered to be the result of aberrant wound-healing process that leads to fibrotic scarring, originating from different etiology. Although iatrogenic etiology is the main cause of subglottic or tracheal stenosis, also autoimmune and infectious diseases may be involved in causing LTS. Furthermore, fibrotic obstruction in the anatomic region under the glottis can also be diagnosed without apparent etiology after a comprehensive workup; in this case, the pathological process is called idiopathic subglottic stenosis (iSGS). So far, the laryngotracheal scar resulting from airway injury due to different diseases was considered as inert tissue requiring surgical removal to restore airway patency. However, this assumption has recently been revised by regarding the tracheal scarring process as a fibroinflammatory event due to immunological alteration, similar to other fibrotic diseases. Recent acquisitions suggest that different factors, such as growth factors, cytokines, altered fibroblast function and genetic susceptibility, can all interact in a complex way leading to aberrant and fibrotic wound healing after an insult that acts as a trigger. However, also physiological derangement due to LTS could play a role in promoting dysregulated response to laryngo-tracheal mucosal injury, through biomechanical stress and mechanotransduction activation. The aim of this narrative review is to present the state-of-the-art knowledge regarding molecular mechanisms, as well as mechanical and physio-pathological features behind LTS.


2022 - Phenotypic, functional, and metabolic heterogeneity of immune cells infiltrating non–small cell lung cancer [Articolo su rivista]
Aramini, B.; Masciale, V.; Samarelli, A. V.; Dubini, A.; Gaudio, M.; Stella, F.; Morandi, U.; Dominici, M.; De Biasi, S.; Gibellini, L.; Cossarizza, A.
abstract

Lung cancer is the leading cancer in the world, accounting for 1.2 million of new cases annually, being responsible for 17.8% of all cancer deaths. In particular, non-small cell lung cancer (NSCLC) is involved in approximately 85% of all lung cancers with a high lethality probably due to the asymptomatic evolution, leading patients to be diagnosed when the tumor has already spread to other organs. Despite the introduction of new therapies, which have improved the long-term survival of these patients, this disease is still not well cured and under controlled. Over the past two decades, single-cell technologies allowed to deeply profile both the phenotypic and metabolic aspects of the immune cells infiltrating the TME, thus fostering the identification of predictive biomarkers of prognosis and supporting the development of new therapeutic strategies. In this review, we discuss phenotypic and functional characteristics of the main subsets of tumor-infiltrating lymphocytes (TILs) and tumor-infiltrating myeloid cells (TIMs) that contribute to promote or suppress NSCLC development and progression. We also address two emerging aspects of TIL and TIM biology, i.e., their metabolism, which affects their effector functions, proliferation, and differentiation, and their capacity to interact with cancer stem cells.


2022 - The role of immune response in the pathogenesis of idiopathic pulmonary fibrosis: far beyond the Th1/Th2 imbalance. [Articolo su rivista]
Spagnolo, Paolo; Tonelli, Roberto; Samarelli, ANNA VALERIA; Castelli, Gioele; Cocconcelli, Elisabetta; Petrarulo, Simone; Cerri, Stefania; Bernardinello, Nicol; Clini, Enrico; Saetta, Marina; Balestro., Elisabetta
abstract

Introduction: . Idiopathic pulmonary fibrosis (IPF) is a chronic disease of unknown origin characterized by progressive scarring of the lung leading to irreversible loss of function. Despite the availability of two drugs that are able to slow down disease progression, IPF remains a deadly disease. The pathogenesis of IPF is poorly understood, but a dysregulated wound healing response following recurrent alveolar epithelial injury is thought to be crucial. Areas covered. In the last few years, the role of the immune system in IPF pathobiology has been reconsidered; indeed, recent data suggest that a dysfunctional immune system may promote and unfavorable interplay with pro-fibrotic pathways thus acting as a cofactor in disease development and progression. In this article, we review and critically discuss the role of T cells in the pathogenesis and progression of IPF in the attempt to highlight ways in which further research in this area may enable the development of targeted immunomodulatory therapies for this dreadful disease. Expert opinion: A better understanding of T cells interactions has the potential to facilitate the development of immune modulators targeting multiple T cell-mediated pathways thus halting disease initiation and progression.


2021 - Fibrotic idiopathic interstitial lung disease: the molecular and cellular key players. [Articolo su rivista]
Samarelli, A; Tonelli, R; Marchioni, A; Bruzzi, G; Gozzi, F; Andrisani, D; Castaniere, I; Manicardi, L; Moretti, A; Tabbì, L; Cerri, S; Beghe', B; Dominici, M; Clini, E.
abstract

Interstitial lung disease (ILDs) that are known as diffuse parenchymal lung diseases (DPLDs) lead to the damage of alveolar epithelium and lung parenchyma culminating into inflammation and widespread fibrosis. ILDs that account for more than 200 different pathologies, can be di-vided into two groups: ILDs that have a known cause and those where the cause is unknown clas-sified as Idiopathic Interstitial Pneumonia (IIPs). IIPs include idiopathic pulmonary fibrosis (IPF), non-specific interstitial pneumonia (NSIP), cryptogenic organizing pneumonia (COP) known also as bronchiolitis obliterans organizing pneumonia (BOOP), Acute interstitial pneumonia (AIP), Desquamative Interstitial Pneumonia (DIP), Respiratory bronchiolitis-associated interstitial lung disease (RB-ILD), and lymphocytic interstitial pneumonia (LIP). In this review our aim is to de-scribe the pathogenic mechanisms that lead to the onset and progression of the different IIPs, starting from IPF as the most studied, in order to find both common and standalone molecular and cellular key players among them. Finally, a deeper molecular and cellular characterization of different interstitial lung disease without known cause, would contribute to give a more accurate diagnosis to the patients, that would translate in a more effective treatment decision.


2021 - Mesenchymal stem cell immunomodulation: In pursuit of controlling COVID-19 related cytokine storm [Articolo su rivista]
Song, N.; Wakimoto, H.; Rossignoli, F.; Bhere, D.; Ciccocioppo, R.; Chen, K. -S.; Khalsa, J. K.; Mastrolia, I.; Samarelli, A. V.; Dominici, M.; Shah, K.
abstract

The Coronavirus disease 2019 (COVID-19) pandemic has grown to be a global public health crisis with no safe and effective treatments available yet. Recent findings suggest that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the coronavirus pathogen that causes COVID-19, could elicit a cytokine storm that drives edema, dysfunction of the airway exchange, and acute respiratory distress syndrome in the lung, followed by acute cardiac injury and thromboembolic events leading to multiorgan failure and death. Mesenchymal stem cells (MSCs), owing to their powerful immunomodulatory abilities, have the potential to attenuate the cytokine storm and have therefore been proposed as a potential therapeutic approach for which several clinical trials are underway. Given that intravenous infusion of MSCs results in a significant trapping in the lung, MSC therapy could directly mitigate inflammation, protect alveolar epithelial cells, and reverse lung dysfunction by normalizing the pulmonary microenvironment and preventing pulmonary fibrosis. In this review, we present an overview and perspectives of the SARS-CoV-2 induced inflammatory dysfunction and the potential of MSC immunomodulation for the prevention and treatment of COVID-19 related pulmonary disease.


2021 - Molecular mechanisms and cellular contribution from lung fibrosis to lung cancer development. [Articolo su rivista]
Samarelli, ANNA VALERIA; Masciale, Valentina; Aramini, Beatrice; Pamela Colò, Georgina; Tonelli, Roberto; Marchioni, Alessandro; Bruzzi, Giulia; Gozzi, Filippo; Andrisani, Dario; Castaniere, Ivana; Manicardi, Linda; Moretti, Antonio; Tabbì, Luca; Guaitoli, Giorgia; Cerri, Stefania; Dominici, Massimo; Clini, Enrico
abstract

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fibrosing interstitial lung 28 disease (ILD) of unknown etiology, with a median survival of 2-4 years from the time of diagnosis. 29 Although IPF has unknown etiology by definition, there have been identified several risks factors 30 increasing the probability of the onset and progression of the disease in IPF patients such as cigarette 31 smoking and environmental risks factors associated to domestic and occupational exposure. Among 32 them, cigarette smoking together with concomitant emphysema might predispose IPF patients to 33 lung cancer (LC), mostly to non-small cell lung cancer (NSCLC), increasing the risk of lung cancer 34 development. To this purpose, IPF and LC share several cellular and molecular processes driving 35 the progression of both pathologies such as fibroblast transition proliferation and activation, endo- 36 plasmic reticulum stress, oxidative stress, and many genetic and epigenetic markers that predispose 37 the IPF patients to LC development. Nintedanib, a tyrosine-kinase inhibitor, was firstly developed 38 as an anticancer drug and then recognized as an anti-fibrotic agent based on the common target 39 molecular pathway. In this review our aim is to describe the updated studies on common cellular 40 and molecular mechanisms between IPF and lung cancer, whose knowledge might help to find 41 novel therapeutic targets for this disease combination.


2021 - Pulmonary stretch and lung mechanotransduction: Implications for progression in the fibrotic lung [Articolo su rivista]
Marchioni, A; Tonelli, R; Cerri, S; Castaniere, I; Andrisani, D; Gozzi, F; Bruzzi, G; Manicardi, L; Moretti, A; Demurtas, J; Baroncini, S; Andreani, A; Cappiello, G; Busani, S; Fantini, R; Tabbì, L; Samarelli, A; Clini, E.
abstract

Lung fibrosis results from the synergic interplay between regenerative deficits of the alveolar epithelium and dysregulated mechanisms of repair in response to alveolar and vascular damage, followed by progressive fibroblast and myofibroblast proliferation and excessive deposition of extracellular matrix. The increased parenchymal stiffness of fibrotic lungs significantly affects respiratory mechanics, making the lung more fragile and prone to non-physiological stress during spontaneous breathing and mechanical ventilation. Given their parenchymal inhomogeneity, fibrotic lungs may display an anisotropic response to mechanical stresses with different regional deformations (micro-strain). This behavior is not described by the standard stress-strain curve but follows the mechano-elastic models of “squishy balls”, where the elastic limit can be reached due to the excessive deformation of parenchymal areas with normal elasticity, surrounded by inelastic fibrous tissue or collapsed induration areas, which tend to protrude outside the fibrous ring. Increasing evidence has shown that non-physiological mechanical forces applied to fibrotic lungs with as34 sociated abnormal mechanotransduction could favor the progression of pulmonary fibrosis. With this review we aim at summarizing the state of the art on the relation between mechanical forces acting on the lung and biological response in pulmonary fibrosis, with a focus on the progression of damage in the fibrotic lung during spontaneous breathing and assisted ventilatory support.


2021 - Spontaneous breathing and evolving phenotypes of lung damage in patients with COVID-19. [Articolo su rivista]
Tonelli, R; Marchioni, A; Tabbì, L; Fantini, R; Busani, S; Castaniere, I; Andrisani, D; Gozzi, F; Bruzzi, G; Manicardi, L; Demurtas, J; Andreani, A; Cappiello, G; Samarelli, A; Clini, E.
abstract

The mechanisms of acute respiratory failure other than inflammation and complicating the SARS-CoV-2 infection are still far from being fully understood, thus challenging the management of COVID-19 patients in the critical care setting. In this unforeseen scenario, the role of an individual’s excessive spontaneous breathing may acquire critical importance, being one potential and important driver of lung injury and disease progression. The consequences of this acute lung damage may impair lung structure forecasting the model of a fragile respiratory system. This perspective article aims to analyze the progression of injured lung phenotypes across the SARS-CoV-2 induced respiratory failure, pointing out the role of spontaneous breathing and also tackling the specific respiratory/ventilatory strategy required by the fragile lung type.


2019 - Challenges in Clinical Development of Mesenchymal Stromal/Stem Cells: Concise Review [Articolo su rivista]
Mastrolia, I.; Foppiani, E. M.; Murgia, A.; Candini, O.; Samarelli, A. V.; Grisendi, G.; Veronesi, E.; Horwitz, E. M.; Dominici, M.
abstract

Identified 50 years ago, mesenchymal stromal/stem cells (MSCs) immediately generated a substantial interest among the scientific community because of their differentiation plasticity and hematopoietic supportive function. Early investigations provided evidence of a relatively low engraftment rate and a transient benefit for challenging congenital and acquired diseases. The reasons for these poor therapeutic benefits forced the entire field to reconsider MSC mechanisms of action together with their ex vivo manipulation procedures. This phase resulted in advances in MSCs processing and the hypothesis that MSC-tissue supportive functions may be prevailing their differentiation plasticity, broadening the spectrum of MSCs therapeutic potential far beyond their lineage-restricted commitments. Consequently, an increasing number of studies have been conducted for a variety of clinical indications, revealing additional challenges and suggesting that MSCs are still lagging behind for a solid clinical translation. For this reason, our aim was to dissect the current challenges in the development of still promising cell types that, after more than half a century, still need to reach their maturity. Stem Cells Translational Medicine 2019;8:1135–1148.


2019 - Impact of HOXB7 overexpression on human adipose-derived mesenchymal progenitors [Articolo su rivista]
Foppiani, E. M.; Candini, O.; Mastrolia, I.; Murgia, A.; Grisendi, G.; Samarelli, A. V.; Boscaini, G.; Pacchioni, L.; Pinelli, M.; De Santis, G.; Horwitz, E. M.; Veronesi, E.; Dominici, M.
abstract

Background: The ex vivo expansion potential of mesenchymal stromal/stem cells (MSC) together with their differentiation and secretion properties makes these cells an attractive tool for transplantation and tissue engineering. Although the use of MSC is currently being tested in a growing number of clinical trials, it is still desirable to identify molecular markers that may help improve their performance both in vitro and after transplantation. Methods: Recently, HOXB7 was identified as a master player driving the proliferation and differentiation of bone marrow mesenchymal progenitors. In this study, we investigated the effect of HOXB7 overexpression on the ex vivo features of adipose mesenchymal progenitors (AD-MSC). Results: HOXB7 increased AD-MSC proliferation potential, reduced senescence, and improved chondrogenesis together with a significant increase of basic fibroblast growth factor (bFGF) secretion. Conclusion: While further investigations and in vivo models shall be applied for better understanding, these data suggest that modulation of HOXB7 may be a strategy for innovative tissue regeneration applications.