57 results on '"Bellet MM"'
Search Results
2. Thymosin alpha 1 in cystic fibrosis: from the lung to the gut
- Author
-
Pariano, M, Renga, G, Stincardini, C, Borghi, M, Sellitto, F, Sforna, L, D’Onofrio, F, Costantini, C, Romani, L, and Bellet, Mm
- Published
- 2020
3. Bacteria and fungi of the lung: allies or enemies?
- Author
-
Garaci E, Pariano M, Nunzi E, Costantini C, Bellet MM, Antognelli C, Russo MA, and Romani L
- Abstract
Moving from the earlier periods in which the lungs were believed to represent sterile environments, our knowledge on the lung microbiota has dramatically increased, from the first descriptions of the microbial communities inhabiting the healthy lungs and the definition of the ecological rules that regulate its composition, to the identification of the changes that occur in pathological conditions. Despite the limitations of lung as a microbiome reservoir due to the low microbial biomass and abundance, defining its microbial composition and function in the upper and lower airways may help understanding the impact on local homeostasis and its disruption in lung diseases. In particular, the understanding of the metabolic and immune significance of microbes, their presence or lack thereof, in health and disease states could be valuable in development of novel druggable targets in disease treatments. Next-generation sequencing has identified intricate inter-microbe association networks that comprise true mutualistic or antagonistic direct or indirect relationships in the respiratory tract. In this review, the tripartite interaction of bacteria, fungi and the mammalian host is addressed to provide an integrated view of the microbial-host cross-talk in lung health and diseases from an immune and metabolic perspective., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Garaci, Pariano, Nunzi, Costantini, Bellet, Antognelli, Russo and Romani.)
- Published
- 2024
- Full Text
- View/download PDF
4. Protective Effect of Indole-3-Aldehyde in Murine COVID-19-Associated Pulmonary Aspergillosis.
- Author
-
Pariano M, Gidari A, Stincardini C, Pierucci S, Bastianelli S, Puccetti M, Giovagnoli S, Bellet MM, Fabi C, Castronari R, Antognelli C, Costantini C, Ricci M, Francisci D, and Romani L
- Abstract
Aspergillus fumigatus is an environmental fungus recently included in the fungal high-priority pathogens by the World Health Organization. While immunodeficiency and/or pre-existing lung damage represent a well-recognized fertile ground for fungal growth, it is increasingly being recognized that severe viral infections may similarly favor A. fumigatus colonization and infection, as recently experienced in the Coronavirus disease 2019 (COVID-19) pandemic. Herein, in a murine model of COVID-19-associated pulmonary aspergillosis (CAPA), obtained by the concomitant exposure to the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Spike protein and A. fumigatus conidia, we found that the microbial compound indole-3-aldehyde (3-IAld) was able to ameliorate CAPA by working at multiple levels during viral infection and fungal superinfection, including epithelial barrier protection, promotion of antiviral responses, and limiting viral replication. As a consequence, 3-IAld limited the pathogenic sequelae of fungal superinfection as revealed by the controlled fungal burden and restrained inflammatory pathology. These results point to indole compounds as potential agents to prevent CAPA.
- Published
- 2024
- Full Text
- View/download PDF
5. The immune and microbial homeostasis determines the Candida -mast cells cross-talk in celiac disease.
- Author
-
Renga G, Pariano M, D'Onofrio F, Pieraccini G, Di Serio C, Villella VR, Abbate C, Puccetti M, Giovagnoli S, Stincardini C, Bellet MM, Ricci M, Costantini C, Oikonomou V, and Romani L
- Subjects
- Humans, Gastrointestinal Microbiome immunology, Dysbiosis immunology, Candidiasis immunology, Candidiasis microbiology, Animals, Candida pathogenicity, Candida immunology, Intestinal Mucosa microbiology, Intestinal Mucosa immunology, Intestinal Mucosa metabolism, Celiac Disease immunology, Celiac Disease microbiology, Celiac Disease metabolism, Homeostasis, Candida albicans pathogenicity, Candida albicans immunology, Mast Cells immunology, Mast Cells metabolism
- Abstract
Celiac disease (CD) is an autoimmune enteropathy resulting from an interaction between diet, genome, and immunity. Although many patients respond to a gluten-free diet, in a substantive number of individuals, the intestinal injury persists. Thus, other factors might amplify the ongoing inflammation. Candida albicans is a commensal fungus that is well adapted to the intestinal life. However, specific conditions increase Candida pathogenicity. The hypothesis that Candida may be a trigger in CD has been proposed after the observation of similarity between a fungal wall component and two CD-related gliadin T-cell epitopes. However, despite being implicated in intestinal disorders, Candida may also protect against immune pathologies highlighting a more intriguing role in the gut. Herein, we postulated that a state of chronic inflammation associated with microbial dysbiosis and leaky gut are favorable conditions that promote C. albicans pathogenicity eventually contributing to CD pathology via a mast cells (MC)-IL-9 axis. However, the restoration of immune and microbial homeostasis promotes a beneficial C. albicans -MC cross-talk favoring the attenuation of CD pathology to alleviate CD pathology and symptoms., (© 2024 Renga et al.)
- Published
- 2024
- Full Text
- View/download PDF
6. Author Correction: Bridging of host-microbiota tryptophan partitioning by the serotonin pathway in fungal pneumonia.
- Author
-
Renga G, D'Onofrio F, Pariano M, Galarini R, Barola C, Stincardini C, Bellet MM, Ellemunter H, Lass-Flörl C, Costantini C, Napolioni V, Ehrlich AK, Antognelli C, Fini M, Garaci E, Nunzi E, and Romani L
- Published
- 2024
- Full Text
- View/download PDF
7. Dual species sphingosine-1-phosphate lyase inhibitors to combine antifungal and anti-inflammatory activities in cystic fibrosis: a feasibility study.
- Author
-
Cellini B, Pampalone G, Camaioni E, Pariano M, Catalano F, Zelante T, Dindo M, Macchioni L, Di Veroli A, Galarini R, Paoletti F, Davidescu M, Stincardini C, Vascelli G, Bellet MM, Saba J, Giovagnoli S, Giardina G, Romani L, and Costantini C
- Subjects
- Humans, Animals, Mice, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Antifungal Agents pharmacology, Antifungal Agents therapeutic use, Feasibility Studies, Inflammation, Anti-Inflammatory Agents pharmacology, Anti-Inflammatory Agents therapeutic use, Cystic Fibrosis drug therapy
- Abstract
Cystic fibrosis (CF) is an autosomal recessive disorder characterized by respiratory failure due to a vicious cycle of defective Cystic Fibrosis Transmembrane conductance Regulator (CFTR) function, chronic inflammation and recurrent bacterial and fungal infections. Although the recent introduction of CFTR correctors/potentiators has revolutionized the clinical management of CF patients, resurgence of inflammation and persistence of pathogens still posit a major concern and should be targeted contextually. On the background of a network-based selectivity that allows to target the same enzyme in the host and microbes with different outcomes, we focused on sphingosine-1-phosphate (S1P) lyase (SPL) of the sphingolipid metabolism as a potential candidate to uniquely induce anti-inflammatory and antifungal activities in CF. As a feasibility study, herein we show that interfering with S1P metabolism improved the immune response in a murine model of CF with aspergillosis while preventing germination of Aspergillus fumigatus conidia. In addition, in an early drug discovery process, we purified human and A. fumigatus SPL, characterized their biochemical and structural properties, and performed an in silico screening to identify potential dual species SPL inhibitors. We identified two hits behaving as competitive inhibitors of pathogen and host SPL, thus paving the way for hit-to-lead and translational studies for the development of drug candidates capable of restraining fungal growth and increasing antifungal resistance., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
8. Role of the Circadian Gas-Responsive Hemeprotein NPAS2 in Physiology and Pathology.
- Author
-
Murgo E, Colangelo T, Bellet MM, Malatesta F, and Mazzoccoli G
- Abstract
Neuronal PAS domain protein 2 (NPAS2) is a hemeprotein comprising a basic helix-loop-helix domain (bHLH) and two heme-binding sites, the PAS-A and PAS-B domains. This protein acts as a pyridine nucleotide-dependent and gas-responsive CO-dependent transcription factor and is encoded by a gene whose expression fluctuates with circadian rhythmicity. NPAS2 is a core cog of the molecular clockwork and plays a regulatory role on metabolic pathways, is important for the function of the central nervous system in mammals, and is involved in carcinogenesis as well as in normal biological functions and processes, such as cardiovascular function and wound healing. We reviewed the scientific literature addressing the various facets of NPAS2 and framing this gene/protein in several and very different research and clinical fields.
- Published
- 2023
- Full Text
- View/download PDF
9. Bridging of host-microbiota tryptophan partitioning by the serotonin pathway in fungal pneumonia.
- Author
-
Renga G, D'Onofrio F, Pariano M, Galarini R, Barola C, Stincardini C, Bellet MM, Ellemunter H, Lass-Flörl C, Costantini C, Napolioni V, Ehrlich AK, Antognelli C, Fini M, Garaci E, Nunzi E, and Romani L
- Subjects
- Humans, Animals, Mice, Tryptophan, Serotonin, Pneumonia, Mycoses, Microbiota, Aspergillosis, Influenza, Human
- Abstract
The aromatic amino acid L-tryptophan (Trp) is essentially metabolized along the host and microbial pathways. While much is known about the role played by downstream metabolites of each pathways in intestinal homeostasis, their role in lung immune homeostasis is underappreciated. Here we have examined the role played by the Trp hydroxylase/5-hydroxytryptamine (5-HT) pathway in calibrating host and microbial Trp metabolism during Aspergillus fumigatus pneumonia. We found that 5-HT produced by mast cells essentially contributed to pathogen clearance and immune homeostasis in infection by promoting the host protective indoleamine-2,3-dioxygenase 1/kynurenine pathway and limiting the microbial activation of the indole/aryl hydrocarbon receptor pathway. This occurred via regulation of lung and intestinal microbiota and signaling pathways. 5-HT was deficient in the sputa of patients with Cystic fibrosis, while 5-HT supplementation restored the dysregulated Trp partitioning in murine disease. These findings suggest that 5-HT, by bridging host-microbiota Trp partitioning, may have clinical effects beyond its mood regulatory function in respiratory pathologies with an inflammatory component., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
10. COVID-19 and beyond: Reassessing the role of thymosin alpha1 in lung infections.
- Author
-
Bellet MM, Renga G, Pariano M, Stincardini C, D'Onofrio F, Goldstein AL, Garaci E, Romani L, and Costantini C
- Subjects
- Humans, Thymalfasin therapeutic use, Pandemics, Lung, Thymosin therapeutic use, COVID-19
- Abstract
The recent COVID-19 pandemic has catalyzed the attention of the scientific community to the long-standing issue of lower respiratory tract infections. The myriad of airborne bacterial, viral and fungal agents to which humans are constantly exposed represents a constant threat to susceptible individuals and bears the potential to reach a catastrophic scale when the ease of inter-individual transmission couples with a severe pathogenicity. While we might be past the threat of COVID-19, the risk of future outbreaks of respiratory infections is tangible and argues for a comprehensive assessment of the pathogenic mechanisms shared by airborne pathogens. On this regard, it is clear that the immune system play a major role in dictating the clinical course of the infection. A balanced immune response is required not only to disarm the pathogens, but also to prevent collateral tissue damage, thus moving at the interface between resistance to infection and tolerance. Thymosin alpha1 (Tα1), an endogenous thymic peptide, is increasingly being recognized for its ability to work as an immunoregulatory molecule able to balance a derailed immune response, working as immune stimulatory or immune suppressive in a context-dependent manner. In this review, we will take advantage from the recent work on the COVID-19 pandemic to reassess the role of Tα1 as a potential therapeutic molecule in lung infections caused by either defective or exaggerated immune responses. The elucidation of the immune regulatory mechanisms of Tα1 might open a new window of opportunity for the clinical translation of this enigmatic molecule and a potential new weapon in our arsenal against lung infections., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
11. Aryl Hydrocarbon Receptor Agonism Antagonizes the Hypoxia-driven Inflammation in Cystic Fibrosis.
- Author
-
Pariano M, Puccetti M, Stincardini C, Napolioni V, Gatticchi L, Galarini R, Renga G, Barola C, Bellet MM, D'Onofrio F, Nunzi E, Bartoli A, Antognelli C, Cariani L, Russo M, Porcaro L, Colombo C, Majo F, Lucidi V, Montemitro E, Fiscarelli E, Ellemunter H, Lass-Flörl C, Ricci M, Costantini C, Giovagnoli S, and Romani L
- Subjects
- Humans, Mice, Animals, Hypoxia metabolism, Signal Transduction, Inflammation, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Receptors, Aryl Hydrocarbon metabolism, Cystic Fibrosis
- Abstract
Hypoxia contributes to the exaggerated yet ineffective airway inflammation that fails to oppose infections in cystic fibrosis (CF). However, the potential for impairment of essential immune functions by HIF-1α (hypoxia-inducible factor 1α) inhibition demands a better comprehension of downstream hypoxia-dependent pathways that are amenable for manipulation. We assessed here whether hypoxia may interfere with the activity of AhR (aryl hydrocarbon receptor), a versatile environmental sensor highly expressed in the lungs, where it plays a homeostatic role. We used murine models of Aspergillus fumigatus infection in vivo and human cells in vitro to define the functional role of AhR in CF, evaluate the impact of hypoxia on AhR expression and activity, and assess whether AhR agonism may antagonize hypoxia-driven inflammation. We demonstrated that there is an important interferential cross-talk between the AhR and HIF-1α signaling pathways in murine and human CF, in that HIF-1α induction squelched the normal AhR response through an impaired formation of the AhR:ARNT (aryl hydrocarbon receptor nuclear translocator)/HIF-1β heterodimer. However, functional studies and analysis of the AhR genetic variability in patients with CF proved that AhR agonism could prevent hypoxia-driven inflammation, restore immune homeostasis, and improve lung function. This study emphasizes the contribution of environmental factors, such as infections, in CF disease progression and suggests the exploitation of hypoxia:xenobiotic receptor cross-talk for antiinflammatory therapy in CF.
- Published
- 2023
- Full Text
- View/download PDF
12. The circadian control of tryptophan metabolism regulates the host response to pulmonary fungal infections.
- Author
-
Stincardini C, Pariano M, D'Onofrio F, Renga G, Orecchini E, Orabona C, Nunzi E, Gargaro M, Fallarino F, Chun SK, Fortin BM, Masri S, Brancorsini S, Romani L, Costantini C, and Bellet MM
- Abstract
The environmental light/dark cycle has left its mark on the body's physiological functions to condition not only our inner biology, but also the interaction with external cues. In this scenario, the circadian regulation of the immune response has emerged as a critical factor in defining the host-pathogen interaction and the identification of the underlying circuitry represents a prerequisite for the development of circadian-based therapeutic strategies. The possibility to track down the circadian regulation of the immune response to a metabolic pathway would represent a unique opportunity in this direction. Herein, we show that the metabolism of the essential amino acid tryptophan, involved in the regulation of fundamental processes in mammals, is regulated in a circadian manner in both murine and human cells and in mouse tissues. By resorting to a murine model of pulmonary infection with the opportunistic fungus Aspergillus fumigatus , we showed that the circadian oscillation in the lung of the tryptophan-degrading enzyme indoleamine 2,3-dioxygenase (IDO)1, generating the immunoregulatory kynurenine, resulted in diurnal changes in the immune response and the outcome of fungal infection. In addition, the circadian regulation of IDO1 drives such diurnal changes in a pre-clinical model of cystic fibrosis (CF), an autosomal recessive disease characterized by progressive lung function decline and recurrent infections, thus acquiring considerable clinical relevance. Our results demonstrate that the circadian rhythm at the intersection between metabolism and immune response underlies the diurnal changes in host-fungal interaction, thus paving the way for a circadian-based antimicrobial therapy., (© The Author(s) 2023. Published by Oxford University Press on behalf of National Academy of Sciences.)
- Published
- 2023
- Full Text
- View/download PDF
13. CAR T cells targeting Aspergillus fumigatus are effective at treating invasive pulmonary aspergillosis in preclinical models.
- Author
-
Seif M, Kakoschke TK, Ebel F, Bellet MM, Trinks N, Renga G, Pariano M, Romani L, Tappe B, Espie D, Donnadieu E, Hünniger K, Häder A, Sauer M, Damotte D, Alifano M, White PL, Backx M, Nerreter T, Machwirth M, Kurzai O, Prommersberger S, Einsele H, Hudecek M, and Löffler J
- Subjects
- Animals, Antifungal Agents, Aspergillus fumigatus, Cytokines, Granzymes, Mice, Perforin, T-Lymphocytes, Invasive Pulmonary Aspergillosis therapy, Receptors, Chimeric Antigen
- Abstract
Aspergillus fumigatus is a ubiquitous mold that can cause severe infections in immunocompromised patients, typically manifesting as invasive pulmonary aspergillosis (IPA). Adaptive and innate immune cells that respond to A. fumigatus are present in the endogenous repertoire of patients with IPA but are infrequent and cannot be consistently isolated and expanded for adoptive immunotherapy. Therefore, we gene-engineered A. fumigatus -specific chimeric antigen receptor (Af-CAR) T cells and demonstrate their ability to confer antifungal reactivity in preclinical models in vitro and in vivo. We generated a CAR targeting domain AB90-E8 that recognizes a conserved protein antigen in the cell wall of A. fumigatus hyphae. T cells expressing the Af-CAR recognized A. fumigatus strains and clinical isolates and exerted a direct antifungal effect against A. fumigatus hyphae. In particular, CD8
+ Af-CAR T cells released perforin and granzyme B and damaged A. fumigatus hyphae. CD8+ and CD4+ Af-CAR T cells produced cytokines that activated macrophages to potentiate the antifungal effect. In an in vivo model of IPA in immunodeficient mice, CD8+ Af-CAR T cells localized to the site of infection, engaged innate immune cells, and reduced fungal burden in the lung. Adoptive transfer of CD8+ Af-CAR T cells conferred greater antifungal efficacy compared to CD4+ Af-CAR T cells and an improvement in overall survival. Together, our study illustrates the potential of gene-engineered T cells to treat aggressive infectious diseases that are difficult to control with conventional antimicrobial therapy and support the clinical development of Af-CAR T cell therapy to treat IPA.- Published
- 2022
- Full Text
- View/download PDF
14. The Role of NLRP3 Inflammasome Activation and Oxidative Stress in Varicocele-Mediated Male Hypofertility.
- Author
-
Poli G, Fabi C, Sugoni C, Bellet MM, Costantini C, Luca G, and Brancorsini S
- Subjects
- Antioxidants pharmacology, Humans, Inflammasomes metabolism, Inflammation, Male, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Oxidative Stress, Reactive Oxygen Species pharmacology, Varicocele drug therapy
- Abstract
Varicocele (VC) is the most common abnormality identified in men evaluated for hypofertility. Increased levels of reactive oxygen species (ROS) and reduced antioxidants concentrations are key contributors in varicocele-mediated hypofertility. Moreover, inflammation and alterations in testicular immunity negatively impact male fertility. In particular, NLRP3 inflammasome activation was hypothesized to lead to seminal inflammation, in which the levels of specific cytokines, such as IL-1β and IL-18, are overexpressed. In this review, we described the role played by oxidative stress (OS), inflammation, and NLRP3 inflammasome activation in VC disease. The consequences of ROS overproduction in testis, including inflammation, lipid peroxidation, mitochondrial dysfunction, chromatin damage, and sperm DNA fragmentation, leading to abnormal testicular function and failed spermatogenesis, were highlighted. Finally, we described some therapeutic antioxidant strategies, with recognized beneficial effects in counteracting OS and inflammation in testes, as possible therapeutic drugs against varicocele-mediated hypofertility.
- Published
- 2022
- Full Text
- View/download PDF
15. A High-Risk Profile for Invasive Fungal Infections Is Associated with Altered Nasal Microbiota and Niche Determinants.
- Author
-
Costantini C, Nunzi E, Spolzino A, Merli F, Facchini L, Spadea A, Melillo L, Codeluppi K, Marchesi F, Marchesini G, Valente D, Dragonetti G, Nadali G, Englmaier L, Coufalikova K, Spáčil Z, Bellet MM, Pariano M, Renga G, Stincardini C, D'Onofrio F, Bozza S, Pagano L, Aversa F, and Romani L
- Subjects
- Bacteria, Humans, Nose microbiology, Prospective Studies, Invasive Fungal Infections, Microbiota
- Abstract
It is becoming increasingly clear that the communities of microorganisms that populate the surfaces exposed to the external environment, termed microbiota, are key players in the regulation of pathogen-host cross talk affecting the onset as well as the outcome of infectious diseases. We have performed a multicenter, prospective, observational study in which nasal and oropharyngeal swabs were collected for microbiota predicting the risk of invasive fungal infections (IFIs) in patients with hematological malignancies. Here, we demonstrate that the nasal and oropharyngeal microbiota are different, although similar characteristics differentiate high-risk from low-risk samples at both sites. Indeed, similar to previously published results on the oropharyngeal microbiota, high-risk samples in the nose were characterized by low diversity, a loss of beneficial bacteria, and an expansion of potentially pathogenic taxa, in the presence of reduced levels of tryptophan (Trp). At variance with oropharyngeal samples, however, low Trp levels were associated with defective host-derived kynurenine production, suggesting reduced tolerance mechanisms at the nasal mucosal surface. This was accompanied by reduced levels of the chemokine interleukin-8 (IL-8), likely associated with a reduced recruitment of neutrophils and impaired fungal clearance. Thus, the nasal and pharyngeal microbiomes of hematological patients provide complementary information that could improve predictive tools for the risk of IFI in hematological patients.
- Published
- 2022
- Full Text
- View/download PDF
16. Optimizing therapeutic outcomes of immune checkpoint blockade by a microbial tryptophan metabolite.
- Author
-
Renga G, Nunzi E, Pariano M, Puccetti M, Bellet MM, Pieraccini G, D'Onofrio F, Santarelli I, Stincardini C, Aversa F, Riuzzi F, Antognelli C, Gargaro M, Bereshchenko O, Ricci M, Giovagnoli S, Romani L, and Costantini C
- Subjects
- Animals, Humans, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Mice, Treatment Outcome, Tryptophan pharmacology, Colitis chemically induced, Colitis drug therapy, Neoplasms drug therapy
- Abstract
Background: Despite the great success, the therapeutic benefits of immune checkpoint inhibitors (ICIs) in cancer immunotherapy are limited by either various resistance mechanisms or ICI-associated toxic effects including gastrointestinal toxicity. Thus, novel therapeutic strategies that provide manageable side effects to existing ICIs would enhance and expand their therapeutic efficacy and application. Due to its proven role in cancer development and immune regulation, gut microbiome has gained increasing expectation as a potential armamentarium to optimize immunotherapy with ICI. However, much has to be learned to fully harness gut microbiome for clinical applicability. Here we have assessed whether microbial metabolites working at the interface between microbes and the host immune system may optimize ICI therapy., Methods: To this purpose, we have tested indole-3-carboxaldehyde (3-IAld), a microbial tryptophan catabolite known to contribute to epithelial barrier function and immune homeostasis in the gut via the aryl hydrocarbon receptor (AhR), in different murine models of ICI-induced colitis. Epithelial barrier integrity, inflammation and changes in gut microbiome composition and function were analyzed. AhR, indoleamine 2,3-dioxygenase 1, interleukin (IL)-10 and IL-22 knockout mice were used to investigate the mechanism of 3-IAld activity. The function of the microbiome changes induced by 3-IAld was evaluated on fecal microbiome transplantation (FMT). Finally, murine tumor models were used to assess the effect of 3-IAld treatment on the antitumor activity of ICI., Results: On administration to mice with ICI-induced colitis, 3-IAld protected mice from intestinal damage via a dual action on both the host and the microbes. Indeed, paralleling the activation of the host AhR/IL-22-dependent pathway, 3-IAld also affected the composition and function of the microbiota such that FMT from 3-IAld-treated mice protected against ICI-induced colitis with the contribution of butyrate-producing bacteria. Importantly, while preventing intestinal damage, 3-IAld did not impair the antitumor activity of ICI., Conclusions: This study provides a proof-of-concept demonstration that moving past bacterial phylogeny and focusing on bacterial metabolome may lead to a new class of discrete molecules, and that working at the interface between microbes and the host immune system may optimize ICI therapy., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
17. Anakinra restores cellular proteostasis by coupling mitochondrial redox balance to autophagy.
- Author
-
van de Veerdonk FL, Renga G, Pariano M, Bellet MM, Servillo G, Fallarino F, De Luca A, Iannitti RG, Piobbico D, Gargaro M, Manni G, D'Onofrio F, Stincardini C, Sforna L, Borghi M, Castelli M, Pieroni S, Oikonomou V, Villella VR, Puccetti M, Giovagnoli S, Galarini R, Barola C, Maiuri L, Della Fazia MA, Cellini B, Talesa VN, Dinarello CA, Costantini C, and Romani L
- Subjects
- Animals, Female, Male, Mice, Mice, Knockout, Oxidation-Reduction drug effects, Autophagy drug effects, Interleukin 1 Receptor Antagonist Protein pharmacology, Mitochondria metabolism, Oxidative Stress drug effects, Proteostasis drug effects
- Abstract
Autophagy selectively degrades aggregation-prone misfolded proteins caused by defective cellular proteostasis. However, the complexity of autophagy may prevent the full appreciation of how its modulation could be used as a therapeutic strategy in disease management. Here, we define a molecular pathway through which recombinant IL-1 receptor antagonist (IL-1Ra, anakinra) affects cellular proteostasis independently from the IL-1 receptor (IL-1R1). Anakinra promoted H2O2-driven autophagy through a xenobiotic sensing pathway involving the aryl hydrocarbon receptor that, activated through the indoleamine 2,3-dioxygenase 1-kynurenine pathway, transcriptionally activated NADPH oxidase 4 independent of the IL-1R1. By coupling the mitochondrial redox balance to autophagy, anakinra improved the dysregulated proteostasis network in murine and human cystic fibrosis. We anticipate that anakinra may represent a therapeutic option in addition to its IL-1R1-dependent antiinflammatory properties by acting at the intersection of mitochondrial oxidative stress and autophagy with the capacity to restore conditions in which defective proteostasis leads to human disease.
- Published
- 2022
- Full Text
- View/download PDF
18. A Shifted Composition of the Lung Microbiota Conditions the Antifungal Response of Immunodeficient Mice.
- Author
-
Nunzi E, Renga G, Palmieri M, Pieraccini G, Pariano M, Stincardini C, D'Onofrio F, Santarelli I, Bellet MM, Bartoli A, Costantini C, and Romani L
- Subjects
- Adaptive Immunity, Animals, Aspergillosis microbiology, Aspergillus fumigatus physiology, Fatty Acids, Volatile immunology, Host-Pathogen Interactions, Immunity, Innate, Lung immunology, Mice, Mice, Inbred C57BL, Microbiota, Aspergillosis immunology, Aspergillus fumigatus immunology, Lung microbiology
- Abstract
The microbiome, i.e., the communities of microbes that inhabit the surfaces exposed to the external environment, participates in the regulation of host physiology, including the immune response against pathogens. At the same time, the immune response shapes the microbiome to regulate its composition and function. How the crosstalk between the immune system and the microbiome regulates the response to fungal infection has remained relatively unexplored. We have previously shown that strict anaerobes protect from infection with the opportunistic fungus Aspergillus fumigatus by counteracting the expansion of pathogenic Proteobacteria. By resorting to immunodeficient mouse strains, we found that the lung microbiota could compensate for the lack of B and T lymphocytes in Rag1
-/- mice by skewing the composition towards an increased abundance of protective anaerobes such as Clostridia and Bacteroidota. Conversely, NSG mice, with major defects in both the innate and adaptive immune response, showed an increased susceptibility to infection associated with a low abundance of strict anaerobes and the expansion of Proteobacteria. Further exploration in a murine model of chronic granulomatous disease, a primary form of immunodeficiency characterized by defective phagocyte NADPH oxidase, confirms the association of lung unbalance between anaerobes and Proteobacteria and the susceptibility to aspergillosis. Consistent changes in the lung levels of short-chain fatty acids between the different strains support the conclusion that the immune system and the microbiota are functionally intertwined during Aspergillus infection and determine the outcome of the infection.- Published
- 2021
- Full Text
- View/download PDF
19. Pharyngeal Microbial Signatures Are Predictive of the Risk of Fungal Pneumonia in Hematologic Patients.
- Author
-
Costantini C, Nunzi E, Spolzino A, Palmieri M, Renga G, Zelante T, Englmaier L, Coufalikova K, Spáčil Z, Borghi M, Bellet MM, Acerbi E, Puccetti M, Giovagnoli S, Spaccapelo R, Talesa VN, Lomurno G, Merli F, Facchini L, Spadea A, Melillo L, Codeluppi K, Marchesi F, Marchesini G, Valente D, Dragonetti G, Nadali G, Pagano L, Aversa F, and Romani L
- Subjects
- Animals, Antifungal Agents pharmacology, Antifungal Agents therapeutic use, Disease Models, Animal, Hematologic Neoplasms complications, Humans, Metagenome, Metagenomics methods, Mice, Mycoses diagnosis, Mycoses drug therapy, Pneumonia diagnosis, Pneumonia drug therapy, Risk Assessment, Risk Factors, Hematologic Diseases complications, Microbiota, Mycoses etiology, Pharynx microbiology, Pneumonia etiology
- Abstract
The ability to predict invasive fungal infections (IFI) in patients with hematological malignancies is fundamental for successful therapy. Although gut dysbiosis is known to occur in hematological patients, whether airway dysbiosis also contributes to the risk of IFI has not been investigated. Nasal and oropharyngeal swabs were collected for functional microbiota characterization in 173 patients with hematological malignancies recruited in a multicenter, prospective, observational study and stratified according to the risk of developing IFI. A lower microbial richness and evenness were found in the pharyngeal microbiota of high-risk patients that were associated with a distinct taxonomic and metabolic profile. A murine model of IFI provided biologic plausibility for the finding that loss of protective anaerobes, such as Clostridiales and Bacteroidetes , along with an apparent restricted availability of tryptophan, is causally linked to the risk of IFI in hematologic patients and indicates avenues for antimicrobial stewardship and metabolic reequilibrium in IFI.
- Published
- 2021
- Full Text
- View/download PDF
20. Indole-3-Carboxaldehyde Restores Gut Mucosal Integrity and Protects from Liver Fibrosis in Murine Sclerosing Cholangitis.
- Author
-
D'Onofrio F, Renga G, Puccetti M, Pariano M, Bellet MM, Santarelli I, Stincardini C, Mosci P, Ricci M, Giovagnoli S, Costantini C, and Romani L
- Subjects
- Animals, Diet, Disease Models, Animal, Inflammation complications, Inflammation pathology, Interleukins metabolism, Intestinal Mucosa drug effects, Liver Cirrhosis complications, Mice, Inbred C57BL, Microbiota drug effects, Pyridines, Receptors, Aryl Hydrocarbon metabolism, Interleukin-22, Mice, Cholangitis, Sclerosing pathology, Indoles pharmacology, Intestinal Mucosa pathology, Liver Cirrhosis pathology
- Abstract
Primary sclerosing cholangitis (PSC) is a long-term liver disease characterized by a progressive course of cholestasis with liver inflammation and fibrosis. Intestinal barrier dysfunction has been implicated in the pathogenesis of PSC. According to the "leaky gut" hypothesis, gut inflammation alters the permeability of the intestinal mucosa, with the translocation of gut-derived products that enter the enterohepatic circulation and cause hepatic inflammation. Thus, the administration of molecules that preserve epithelial barrier integrity would represent a promising therapeutic strategy. Indole-3-carboxaldehyde (3-IAld) is a microbial-derived product working at the interface between the host and the microbiota and is able to promote mucosal immune homeostasis in a variety of preclinical settings. Herein, by resorting to a murine model of PSC, we found that 3-IAld formulated for localized delivery in the gut alleviates hepatic inflammation and fibrosis by modulating the intestinal microbiota and activating the aryl hydrocarbon receptor-IL-22 axis to restore mucosal integrity. This study points to the therapeutic potential of 3-IAld in liver pathology.
- Published
- 2021
- Full Text
- View/download PDF
21. Targeted Drug Delivery Technologies Potentiate the Overall Therapeutic Efficacy of an Indole Derivative in a Mouse Cystic Fibrosis Setting.
- Author
-
Puccetti M, Pariano M, Renga G, Santarelli I, D'Onofrio F, Bellet MM, Stincardini C, Bartoli A, Costantini C, Romani L, Ricci M, and Giovagnoli S
- Subjects
- Administration, Inhalation, Aerosols pharmacology, Animals, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Disease Models, Animal, Gastrointestinal Tract drug effects, Gastrointestinal Tract microbiology, Gastrointestinal Tract pathology, Indoles administration & dosage, Indoles pharmacology, Ligands, Lung drug effects, Lung microbiology, Lung pathology, Mice, Inbred C57BL, Receptors, Aryl Hydrocarbon metabolism, Mice, Cystic Fibrosis drug therapy, Cystic Fibrosis pathology, Drug Delivery Systems, Indoles therapeutic use
- Abstract
Inflammation plays a major role in the pathophysiology of cystic fibrosis (CF), a multisystem disease. Anti-inflammatory therapies are, therefore, of interest in CF, provided that the inhibition of inflammation does not compromise the ability to fight pathogens. Here, we assess whether indole-3-aldehyde (3-IAld), a ligand of the aryl hydrocarbon receptor (AhR), may encompass such an activity. We resorted to biopharmaceutical technologies in order to deliver 3-IAld directly into the lung, via dry powder inhalation, or into the gut, via enteric microparticles, in murine models of CF infection and inflammation. We found the site-specific delivery of 3-IAld to be an efficient strategy to restore immune and microbial homeostasis in CF organs, and mitigate lung and gut inflammatory pathology in response to fungal infections, in the relative absence of local and systemic inflammatory toxicity. Thus, enhanced delivery to target organs of AhR agonists, such as 3-IAld, may pave the way for the development of safe and effective anti-inflammatory agents in CF.
- Published
- 2021
- Full Text
- View/download PDF
22. Anakinra Activates Superoxide Dismutase 2 to Mitigate Inflammasome Activity.
- Author
-
Pariano M, Pieroni S, De Luca A, Iannitti RG, Borghi M, Puccetti M, Giovagnoli S, Renga G, D'Onofrio F, Bellet MM, Stincardini C, Della-Fazia MA, Servillo G, van de Veerdonk FL, Costantini C, and Romani L
- Subjects
- Animals, Cells, Cultured, Cystic Fibrosis etiology, Cystic Fibrosis metabolism, Cystic Fibrosis pathology, Disease Models, Animal, Granulomatous Disease, Chronic etiology, Granulomatous Disease, Chronic metabolism, Granulomatous Disease, Chronic pathology, Macrophages, Alveolar metabolism, Macrophages, Alveolar pathology, Mice, Mice, Knockout, Mitochondria metabolism, Oxidation-Reduction, Oxidative Stress, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha metabolism, RAW 264.7 Cells, Reactive Oxygen Species metabolism, Transcription Factors metabolism, Inflammasomes metabolism, Interleukin 1 Receptor Antagonist Protein pharmacology, Recombinant Proteins pharmacology, Superoxide Dismutase metabolism
- Abstract
Inflammasomes are powerful cytosolic sensors of environmental stressors and are critical for triggering interleukin-1 (IL-1)-mediated inflammatory responses. However, dysregulation of inflammasome activation may lead to pathological conditions, and the identification of negative regulators for therapeutic purposes is increasingly being recognized. Anakinra, the recombinant form of the IL-1 receptor antagonist, proved effective by preventing the binding of IL-1 to its receptor, IL-1R1, thus restoring autophagy and dampening NLR family pyrin domain containing 3 (NLRP3) activity. As the generation of mitochondrial reactive oxidative species (ROS) is a critical upstream event in the activation of NLRP3, we investigated whether anakinra would regulate mitochondrial ROS production. By profiling the activation of transcription factors induced in murine alveolar macrophages, we found a mitochondrial antioxidative pathway induced by anakinra involving the manganese-dependent superoxide dismutase (MnSOD) or SOD2. Molecularly, anakinra promotes the binding of SOD2 with the deubiquitinase Ubiquitin Specific Peptidase 36 (USP36) and Constitutive photomorphogenesis 9 (COP9) signalosome, thus increasing SOD2 protein longevity. Functionally, anakinra and SOD2 protects mice from pulmonary oxidative inflammation and infection. On a preclinical level, anakinra upregulates SOD2 in murine models of chronic granulomatous disease (CGD) and cystic fibrosis (CF). These data suggest that protection from mitochondrial oxidative stress may represent an additional mechanism underlying the clinical benefit of anakinra and identifies SOD2 as a potential therapeutic target.
- Published
- 2021
- Full Text
- View/download PDF
23. The Circadian Protein PER1 Modulates the Cellular Response to Anticancer Treatments.
- Author
-
Bellet MM, Stincardini C, Costantini C, Gargaro M, Pieroni S, Castelli M, Piobbico D, Sassone-Corsi P, Della-Fazia MA, Romani L, and Servillo G
- Subjects
- A549 Cells, Animals, Antineoplastic Agents pharmacology, Apoptosis drug effects, Cell Cycle drug effects, Cell Line, Tumor, Circadian Rhythm drug effects, Cisplatin pharmacology, Docetaxel pharmacology, Drug Chronotherapy, Etoposide pharmacology, Humans, Mice, Neoplasms pathology, Neoplasms therapy, Xenograft Model Antitumor Assays, Carcinogenesis genetics, Neoplasms genetics, Period Circadian Proteins genetics, Tumor Suppressor Protein p53 genetics
- Abstract
The circadian clock driven by the daily light-dark and temperature cycles of the environment regulates fundamental physiological processes and perturbations of these sophisticated mechanisms may result in pathological conditions, including cancer. While experimental evidence is building up to unravel the link between circadian rhythms and tumorigenesis, it is becoming increasingly apparent that the response to antitumor agents is similarly dependent on the circadian clock, given the dependence of each drug on the circadian regulation of cell cycle, DNA repair and apoptosis. However, the molecular mechanisms that link the circadian machinery to the action of anticancer treatments is still poorly understood, thus limiting the application of circadian rhythms-driven pharmacological therapy, or chronotherapy, in the clinical practice. Herein, we demonstrate the circadian protein period 1 (PER1) and the tumor suppressor p53 negatively cross-regulate each other's expression and activity to modulate the sensitivity of cancer cells to anticancer treatments. Specifically, PER1 physically interacts with p53 to reduce its stability and impair its transcriptional activity, while p53 represses the transcription of PER1. Functionally, we could show that PER1 reduced the sensitivity of cancer cells to drug-induced apoptosis, both in vitro and in vivo in NOD scid gamma (NSG) mice xenotransplanted with a lung cancer cell line. Therefore, our results emphasize the importance of understanding the relationship between the circadian clock and tumor regulatory proteins as the basis for the future development of cancer chronotherapy.
- Published
- 2021
- Full Text
- View/download PDF
24. Thymosin alpha 1 exerts beneficial extrapulmonary effects in cystic fibrosis.
- Author
-
Bellet MM, Borghi M, Pariano M, Renga G, Stincardini C, D'Onofrio F, Brancorsini S, Garaci E, Costantini C, and Romani L
- Subjects
- Animals, Candida albicans drug effects, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Disease Models, Animal, Female, Homeostasis drug effects, Humans, Immunologic Factors pharmacology, Kynurenine metabolism, Liver, Lung, Mice, Mutation, Obesity drug therapy, Pancreas, Signal Transduction, Thymalfasin pharmacology, Cystic Fibrosis drug therapy, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Immunologic Factors chemistry, Thymalfasin chemistry
- Abstract
Cystic fibrosis (CF) is an autosomal recessive disorder caused by mutations in the gene encoding for the ion channel Cystic Fibrosis Transmembrane conductance Regulator (CFTR). Long considered a lung disease for the devastating impact on the respiratory function, the recent diagnostic and therapeutic advances have shed the light on the extra-pulmonary manifestations of CF, including gastrointestinal, hepatobiliary and pancreatic symptoms. We have previously demonstrated that thymosin alpha1 (Tα1), a naturally occurring immunomodulatory peptide, displays multi-sided beneficial effects in CF that concur in ameliorating the lung inflammatory pathology. In the present study, by resorting to murine models of gut inflammation with clinical relevance for CF patients, we demonstrate that Tα1 can also have beneficial effects in extrapulmonary pathology. Specifically, Tα1 restored barrier integrity and immune homeostasis in the inflamed gut of CF mice as well as in mice with the metabolic syndrome, a disorder that may arise in CF patients with high caloric intake despite pancreatic sufficiency. The protective effects of Tα1 also extended to pancreas and liver, further emphasizing the beneficial effects of Tα1 in extra-pulmonary complications of CF. By performing wide-ranging multi-organ anti-inflammatory effects, Tα1 could potentially integrate current therapeutic approaches to tackle the complex symptomatology of CF disease., Competing Interests: Declaration of competing interest A patent application by L.R. and E.G. on "Thymosin alpha1 for use in the treatment of cystic fibrosis" is pending (International Application Number PCT/IT2016/000027, International Publication Number WO2016/129005 A1)., (Copyright © 2020. Published by Elsevier Masson SAS.)
- Published
- 2021
- Full Text
- View/download PDF
25. Selectively targeting key inflammatory pathways in cystic fibrosis.
- Author
-
Costantini C, Puccetti M, Pariano M, Renga G, Stincardini C, D'Onofrio F, Bellet MM, Cellini B, Giovagnoli S, and Romani L
- Subjects
- Animals, Cystic Fibrosis complications, Cystic Fibrosis metabolism, Cystic Fibrosis pathology, Drug Discovery, Humans, Inflammation complications, Lung drug effects, Lung metabolism, Lung pathology, Cystic Fibrosis drug therapy, Molecular Targeted Therapy methods
- Abstract
Cystic fibrosis (CF) is a rare genetic disorder caused by a defect in the ion channel Cystic Fibrosis Transmembrane conductance Regulator (CFTR), resulting in ionic imbalance of surface fluid. Although affecting multiple organs, the progressive deterioration of respiratory function by recurrent infections and chronic inflammation represents the main cause of morbidity and mortality in CF patients. The development of modulators targeting the basic defect of CFTR has represented a major breakthrough in CF therapy, but the impact on inflammation has remained enigmatic. The emerging scenario taking hold in the field points to inflammation as a major, somehow missed, therapeutic target for prevention of lung decline. Not surprisingly, the development of anti-inflammatory drugs is taking its share in the drug development pipeline. But the path is not straightforward and targeting inflammation should be balanced with the increased risk of infection. The strategy to restore the homeostatic regulation of inflammation to efficiently respond to infection while preventing lung damage needs to be based on identifying and targeting endogenous immunoregulatory pathways that are defective in CF. We herein provide an overview of anti-inflammatory drugs currently approved or under investigation in CF patients, and present our recent studies on how the knowledge on defective immune pathways in CF may translate into innovative and selective anti-inflammatory therapeutics. Through the discovery of naturally occurring molecules or their synthetic mimics, this review emphasizes the critical importance of selectively targeting key inflammatory pathways to preserve immunocompetence in CF patients., Competing Interests: Declaration of competing interest A patent application by L.R. on "Thymosin alpha1 for use in the treatment of cystic fibrosis" is pending (International Application Number PCT/IT2016/000027, International Publication Number WO2016/129005 A1)., (Copyright © 2020 Elsevier Masson SAS. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
26. Editorial: Circadian Rhythm: From Microbes to Hosts.
- Author
-
Bellet MM, Eckel-Mahan K, and Romani L
- Subjects
- Circadian Rhythm, Host-Parasite Interactions
- Published
- 2020
- Full Text
- View/download PDF
27. HOPS/Tmub1 involvement in the NF-kB-mediated inflammatory response through the modulation of TRAF6.
- Author
-
Bellet MM, Pieroni S, Castelli M, Piobbico D, Fallarino F, Romani L, Della-Fazia MA, and Servillo G
- Subjects
- Cell Nucleus metabolism, Humans, Intracellular Signaling Peptides and Proteins genetics, Membrane Proteins genetics, Protein Binding physiology, Signal Transduction physiology, Tumor Suppressor Proteins metabolism, Inflammation metabolism, Intracellular Signaling Peptides and Proteins metabolism, Membrane Proteins metabolism, NF-kappa B metabolism, Transcription Factor RelA metabolism
- Abstract
HOPS/Tmub1 is a ubiquitously expressed transmembrane ubiquitin-like protein that shuttles between nucleus and cytoplasm during cell cycle progression. HOPS causes cell cycle arrest in G
0 /G1 phase, an event associated to stabilization of p19Arf , an important tumor suppressor protein. Moreover, HOPS plays an important role in driving centrosomal assembly and maintenance, mitotic spindle proper organization, and ultimately a correct cell division. Recently, HOPS has been described as an important regulator of p53, which acts as modifier, stabilizing p53 half-life and playing a key role in p53 mediating apoptosis after DNA damage. NF-κB is a transcription factor with a central role in many cellular events, including inflammation and apoptosis. Our experiments demonstrate that the transcriptional activity of the p65/RelA NF-κB subunit is regulated by HOPS. Importantly, Hops-/- cells have remarkable alterations of pro-inflammatory responses. Specifically, we found that HOPS enhances NF-κB activation leading to increase transcription of inflammatory mediators, through the reduction of IκBα stability. Notably, this effect is mediated by a direct HOPS binding to the E3 ubiquitin ligase TRAF6, which lessens TRAF6 stability ultimately leading increased IKK complex activation. These findings uncover a previously unidentified function of HOPS/Tmub1 as a novel modulator of TRAF6, regulating inflammatory responses driven by activation of the NF-κB signaling pathway. The comprehension on how HOPS/Tmub1 takes part to the inflammatory processes in vivo and whether this function is important in the control of proliferation and tumorigenesis could establish the basis for the development of novel pharmacological strategies.- Published
- 2020
- Full Text
- View/download PDF
28. Thymosin α1 protects from CTLA-4 intestinal immunopathology.
- Author
-
Renga G, Bellet MM, Pariano M, Gargaro M, Stincardini C, D'Onofrio F, Mosci P, Brancorsini S, Bartoli A, Goldstein AL, Garaci E, Romani L, and Costantini C
- Subjects
- Animals, CTLA-4 Antigen immunology, CTLA-4 Antigen metabolism, Cell Differentiation drug effects, Dendritic Cells metabolism, Female, Immune Checkpoint Inhibitors adverse effects, Immune Checkpoint Inhibitors pharmacology, Immune Tolerance drug effects, Indoleamine-Pyrrole 2,3,-Dioxygenase metabolism, Intestinal Mucosa metabolism, Intestines drug effects, Male, Mice, Mice, Inbred C57BL, T-Lymphocytes, Regulatory metabolism, Thymalfasin physiology, Thymosin metabolism, Thymosin physiology, Intestinal Mucosa drug effects, Thymalfasin metabolism, Thymalfasin pharmacology
- Abstract
The advent of immune checkpoint inhibitors has represented a major boost in cancer therapy, but safety concerns are increasingly being recognized. Indeed, although beneficial at the tumor site, unlocking a safeguard mechanism of the immune response may trigger autoimmune-like effects at the periphery, thus making the safety of immune checkpoint inhibitors a research priority. Herein, we demonstrate that thymosin α1 (Tα1), an endogenous peptide with immunomodulatory activities, can protect mice from intestinal toxicity in a murine model of immune checkpoint inhibitor-induced colitis. Specifically, Tα1 efficiently prevented immune adverse pathology in the gut by promoting the indoleamine 2,3-dioxygenase (IDO) 1-dependent tolerogenic immune pathway. Notably, Tα1 did not induce IDO1 in the tumor microenvironment, but rather modulated the infiltration of T-cell subsets by inverting the ratio between CD8
+ and Treg cells, an effect that may depend on Tα1 ability to regulate the differentiation and chemokine expression profile of DCs. Thus, through distinct mechanisms that are contingent upon the context, Tα1 represents a plausible candidate to improve the safety/efficacy profile of immune checkpoint inhibitors., (© 2020 Renga et al.)- Published
- 2020
- Full Text
- View/download PDF
29. Pyridoxal 5'-Phosphate-Dependent Enzymes at the Crossroads of Host-Microbe Tryptophan Metabolism.
- Author
-
Cellini B, Zelante T, Dindo M, Bellet MM, Renga G, Romani L, and Costantini C
- Subjects
- Animals, Bacteria metabolism, Humans, Mammals metabolism, Vitamin B 6 metabolism, Host-Pathogen Interactions, Pyridoxal Phosphate metabolism, Tryptophan metabolism
- Abstract
The chemical processes taking place in humans intersects the myriad of metabolic pathways occurring in commensal microorganisms that colonize the body to generate a complex biochemical network that regulates multiple aspects of human life. The role of tryptophan (Trp) metabolism at the intersection between the host and microbes is increasingly being recognized, and multiple pathways of Trp utilization in either direction have been identified with the production of a wide range of bioactive products. It comes that a dysregulation of Trp metabolism in either the host or the microbes may unbalance the production of metabolites with potential pathological consequences. The ability to redirect the Trp flux to restore a homeostatic production of Trp metabolites may represent a valid therapeutic strategy for a variety of pathological conditions, but identifying metabolic checkpoints that could be exploited to manipulate the Trp metabolic network is still an unmet need. In this review, we put forward the hypothesis that pyridoxal 5'-phosphate (PLP)-dependent enzymes, which regulate multiple pathways of Trp metabolism in both the host and in microbes, might represent critical nodes and that modulating the levels of vitamin B6, from which PLP is derived, might represent a metabolic checkpoint to re-orienteer Trp flux for therapeutic purposes.
- Published
- 2020
- Full Text
- View/download PDF
30. Epigenetic Mechanisms of Inflammasome Regulation.
- Author
-
Poli G, Fabi C, Bellet MM, Costantini C, Nunziangeli L, Romani L, and Brancorsini S
- Subjects
- Animals, Disease genetics, Humans, Inflammasomes metabolism, RNA, Untranslated genetics, RNA, Untranslated metabolism, Epigenesis, Genetic, Inflammasomes genetics
- Abstract
The innate immune system represents the host's first-line defense against pathogens, dead cells or environmental factors. One of the most important inflammatory pathways is represented by the activation of the NOD-like receptor (NLR) protein family. Some NLRs induce the assembly of large caspase-1-activating complexes called inflammasomes. Different types of inflammasomes have been identified that can respond to distinct bacterial, viral or fungal infections; sterile cell damage or other stressors, such as metabolic imbalances. Epigenetic regulation has been recently suggested to provide a complementary mechanism to control inflammasome activity. This regulation can be exerted through at least three main mechanisms, including CpG DNA methylation, histones post-translational modifications and noncoding RNA expression. The repression or promotion of expression of different inflammasomes (NLRP1, NLRP2, NLRP3, NLRP4, NLRP6, NLRP7, NLRP12 and AIM2) through epigenetic mechanisms determines the development of pathologies with variable severity. For example, our team recently explored the role of microRNAs (miRNAs) targeting and modulating the components of the inflammasome as potential biomarkers in bladder cancer and during therapy. This suggests that the epigenetic control of inflammasome-related genes could represent a potential target for further investigations of molecular mechanisms regulating inflammatory pathways.
- Published
- 2020
- Full Text
- View/download PDF
31. Rapidly expanded partially HLA DRB1-matched fungus-specific T cells mediate in vitro and in vivo antifungal activity.
- Author
-
Castellano-González G, McGuire HM, Luciani F, Clancy LE, Li Z, Avdic S, Hughes B, Singh M, Fazekas de St Groth B, Renga G, Pariano M, Bellet MM, Romani L, and Gottlieb DJ
- Subjects
- Animals, Antigen-Presenting Cells, Fungi, HLA-DRB1 Chains, Humans, Mice, Antifungal Agents pharmacology, Antifungal Agents therapeutic use, Hematopoietic Stem Cell Transplantation
- Abstract
Invasive fungal infections are a major cause of disease and death in immunocompromised hosts, including patients undergoing allogeneic hematopoietic stem cell transplant (HSCT). Recovery of adaptive immunity after HSCT correlates strongly with recovery from fungal infection. Using initial selection of lymphocytes expressing the activation marker CD137 after fungal stimulation, we rapidly expanded a population of mainly CD4+ T cells with potent antifungal characteristics, including production of tumor necrosis factor α, interferon γ, interleukin-17, and granulocyte-macrophage colony stimulating factor. Cells were manufactured using a fully good manufacturing practice-compliant process. In vitro, the T cells responded to fungal antigens presented on fully and partially HLA-DRB1 antigen-matched presenting cells, including when the single common DRB1 antigen was allelically mismatched. Administration of antifungal T cells lead to reduction in the severity of pulmonary and cerebral infection in an experimental mouse model of Aspergillus. These data support the establishment of a bank of cryopreserved fungus-specific T cells using normal donors with common HLA DRB1 molecules and testing of partially HLA-matched third-party donor fungus-specific T cells as a potential therapeutic in patients with invasive fungal infection after HSCT., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
32. Microbes in the Era of Circadian Medicine.
- Author
-
Costantini C, Renga G, Sellitto F, Borghi M, Stincardini C, Pariano M, Zelante T, Chiarotti F, Bartoli A, Mosci P, Romani L, Brancorsini S, and Bellet MM
- Subjects
- Circadian Rhythm, Humans, Immune System, Symbiosis, Circadian Clocks, Microbiota
- Abstract
The organisms of most domains of life have adapted to circadian changes of the environment and regulate their behavior and physiology accordingly. A particular case of such paradigm is represented by some types of host-pathogen interaction during infection. Indeed, not only some hosts and pathogens are each endowed with their own circadian clock, but they are also influenced by the circadian changes of the other with profound consequences on the outcome of the infection. It comes that daily fluctuations in the availability of resources and the nature of the immune response, coupled with circadian changes of the pathogen, may influence microbial virulence, level of colonization and damage to the host, and alter the equilibrium between commensal and invading microorganisms. In the present review, we discuss the potential relevance of circadian rhythms in human bacterial and fungal pathogens, and the consequences of circadian changes of the host immune system and microbiome on the onset and development of infection. By looking from the perspective of the interplay between host and microbes circadian rhythms, these concepts are expected to change the way we approach human infections, not only by predicting the outcome of the host-pathogen interaction, but also by indicating the best time for intervention to potentiate the anti-microbial activities of the immune system and to weaken the pathogen when its susceptibility is higher., (Copyright © 2020 Costantini, Renga, Sellitto, Borghi, Stincardini, Pariano, Zelante, Chiarotti, Bartoli, Mosci, Romani, Brancorsini and Bellet.)
- Published
- 2020
- Full Text
- View/download PDF
33. Tryptophan Co-Metabolism at the Host-Pathogen Interface.
- Author
-
Costantini C, Bellet MM, Renga G, Stincardini C, Borghi M, Pariano M, Cellini B, Keller N, Romani L, and Zelante T
- Subjects
- Animals, Immunologic Factors physiology, Kynurenine metabolism, Microbiota, Host Microbial Interactions, Tryptophan metabolism
- Published
- 2020
- Full Text
- View/download PDF
34. To Be or Not to Be a Pathogen: Candida albicans and Celiac Disease.
- Author
-
Renga G, Bellet MM, Stincardini C, Pariano M, Oikonomou V, Villella VR, Brancorsini S, Clerici C, Romani L, and Costantini C
- Subjects
- Animals, Biomarkers, Candidiasis immunology, Candidiasis microbiology, Celiac Disease metabolism, Celiac Disease pathology, Cytokines metabolism, Humans, Intestinal Mucosa immunology, Intestinal Mucosa metabolism, Mast Cells immunology, Mast Cells metabolism, Mast Cells pathology, Metabolic Networks and Pathways, Tryptophan metabolism, Candida albicans immunology, Candidiasis complications, Celiac Disease etiology, Disease Susceptibility, Host-Pathogen Interactions
- Abstract
Celiac disease (CD) is an immune-mediated disorder triggered by the ingestion of gluten and characterized by reversible small-bowel mucosal atrophy in genetically predisposed subjects. Although the prevalence of CD has increased, many aspects of this pathology are still unrecognized. Candida albicans , a commensal of the human gastrointestinal tract, has been linked to CD for a long time based, among others, upon the observation of similarity between the fungal wall component, hyphal wall protein 1, and CD-related gliadin T-cell epitopes. We have recently demonstrated that Candida may switch from commensal to pathogen contingent upon several players, including mast cells, key sentinels of the immune system at the interface between the environment and the host, and the pleiotropic cytokine IL-9. However, other factors are likely to play a role by altering the balance between inflammation and tolerance. In this regard, tryptophan and its metabolites are increasingly being recognized in promoting mucosal homeostasis by balancing the immune response to external cues. Based on these premises, we will discuss how the output of Candida colonization in the gut is highly contextual, being determined at the intersection of many immunological (IL-9/mast cells) and metabolic (tryptophan) pathways that ultimately dictate the Candida commensalism vs. pathogenicity in CD, thus paving the way for novel therapeutic opportunities in CD., (Copyright © 2019 Renga, Bellet, Stincardini, Pariano, Oikonomou, Villella, Brancorsini, Clerici, Romani and Costantini.)
- Published
- 2019
- Full Text
- View/download PDF
35. Thymosin β4 promotes autophagy and repair via HIF-1α stabilization in chronic granulomatous disease.
- Author
-
Renga G, Oikonomou V, Moretti S, Stincardini C, Bellet MM, Pariano M, Bartoli A, Brancorsini S, Mosci P, Finocchi A, Rossi P, Costantini C, Garaci E, Goldstein AL, and Romani L
- Subjects
- Actins metabolism, Animals, Autophagy physiology, DNA Repair, Female, Granulomatous Disease, Chronic pathology, Humans, Male, Mice, Mice, Inbred C57BL, NADPH Oxidases metabolism, RAW 264.7 Cells, Granulomatous Disease, Chronic drug therapy, Granulomatous Disease, Chronic metabolism, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Thymosin pharmacology
- Abstract
Chronic granulomatous disease (CGD) is a genetic disorder of the NADPH oxidase characterized by increased susceptibility to infections and hyperinflammation associated with defective autophagy and increased inflammasome activation. Herein, we demonstrate that thymosin β4 (Tβ4), a g-actin sequestering peptide with multiple and diverse intracellular and extracellular activities affecting inflammation, wound healing, fibrosis, and tissue regeneration, promoted in human and murine cells noncanonical autophagy, a form of autophagy associated with phagocytosis and limited inflammation via the death-associated protein kinase 1. We further show that the hypoxia inducible factor-1 (HIF-1)α was underexpressed in CGD but normalized by Tβ4 to promote autophagy and up-regulate genes involved in mucosal barrier protection. Accordingly, inflammation and granuloma formation were impaired and survival increased in CGD mice with colitis or aspergillosis upon Tβ4 treatment or HIF-1α stabilization. Thus, the promotion of endogenous pathways of inflammation resolution through HIF-1α stabilization is druggable in CGD by Tβ4., (© 2019 Renga et al.)
- Published
- 2019
- Full Text
- View/download PDF
36. Targeting the Aryl Hydrocarbon Receptor With Indole-3-Aldehyde Protects From Vulvovaginal Candidiasis via the IL-22-IL-18 Cross-Talk.
- Author
-
Borghi M, Pariano M, Solito V, Puccetti M, Bellet MM, Stincardini C, Renga G, Vacca C, Sellitto F, Mosci P, Brancorsini S, Romani L, and Costantini C
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors immunology, Female, Inflammasomes genetics, Inflammasomes immunology, Interleukin-18 genetics, Interleukins genetics, Mice, Mice, Knockout, NLR Family, Pyrin Domain-Containing 3 Protein genetics, NLR Family, Pyrin Domain-Containing 3 Protein immunology, Receptors, Aryl Hydrocarbon genetics, Receptors, Aryl Hydrocarbon immunology, Signal Transduction genetics, Signal Transduction immunology, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory pathology, Th17 Cells immunology, Th17 Cells pathology, Interleukin-22, Basic Helix-Loop-Helix Transcription Factors agonists, Candida albicans immunology, Candidiasis, Vulvovaginal drug therapy, Candidiasis, Vulvovaginal genetics, Candidiasis, Vulvovaginal immunology, Candidiasis, Vulvovaginal pathology, Indoles pharmacology, Interleukin-18 immunology, Interleukins immunology, Receptors, Aryl Hydrocarbon agonists, Signal Transduction drug effects
- Abstract
Vulvovaginal candidiasis (VVC) is a common mucosal infection caused by Candida spp., most frequently by Candida albicans , which may become recurrent and severely impacting the quality of life of susceptible women. Although it is increasingly being recognized that mucosal damage is mediated by an exaggerated inflammatory response, current therapeutic approaches are only based on antifungals that may relieve the symptomatology, but fail to definitely prevent recurrences. The unrestrained activation of the NLRP3 inflammasome with continuous production of IL-1β and recruitment of neutrophils is recognized as a pathogenic factor in VVC. We have previously shown that IL-22 is required to dampen pathogenic inflammasome activation in VVC via the NLRC4/IL-1Ra axis. However, IL-22 also regulates IL-18, a product of the inflammasome activity that regulates IL-22 expression. Here we describe a cross-regulatory circuit between IL-18 and IL-22 in murine VVC that is therapeutically druggable. We found that IL-18 production was dependent on IL-22 and NLRC4, and that IL-18, in turn, contributes to IL-22 activity. Like in IL-22 deficiency, IL-18 deficiency was associated with an increased susceptibility to VVC and unbalanced Th17/Treg response, suggesting that IL-18 can regulate both the innate and the adaptive responses to the fungus. Administration of the microbial metabolite indole-3-aldehyde, known to stimulate the production of IL-22 via the aryl hydrocarbon receptor (AhR), promoted IL-18 expression and protection against Candida infection. Should low levels of IL-18 be demonstrated in the vaginal fluids of women with recurrent VVC, targeting the AhR/IL-22/IL-18 pathway could be exploited for future therapeutic approaches in VVC. This study suggests that a deeper understanding of the mechanisms regulating inflammasome activity may lead to the identification of novel targets for intervention in VVC., (Copyright © 2019 Borghi, Pariano, Solito, Puccetti, Bellet, Stincardini, Renga, Vacca, Sellitto, Mosci, Brancorsini, Romani and Costantini.)
- Published
- 2019
- Full Text
- View/download PDF
37. A Reappraisal of Thymosin Alpha1 in Cancer Therapy.
- Author
-
Costantini C, Bellet MM, Pariano M, Renga G, Stincardini C, Goldstein AL, Garaci E, and Romani L
- Abstract
Thymosin alpha1 (Tα1), an endogenous peptide first isolated from the thymic tissue in the mid-sixties, has gained considerable attention for its immunostimulatory activity that led to its application to diverse pathological conditions, including cancer. Studies in animal models and human patients have shown promising results in different types of malignancies, especially when Tα1 was used in combination with other chemo- and immune therapies. For this reason, the advancements in our knowledge on the adjuvant role of Tα1 have moved in parallel with the development of novel cancer therapies in a way that Tα1 was integrated to changing paradigms and protocols, and tested for increased efficacy and safety. Cancer immunotherapy has recently experienced a tremendous boost following the development and clinical application of immune checkpoint inhibitors. By unleashing the full potential of the adaptive immune response, checkpoint inhibitors were expected to be very effective against tumors, but it soon became clear that a widespread and successful application was not straightforward and shortcomings in efficacy and safety clearly emerged. This scenario led to the development of novel concepts in immunotherapy and the design of combination protocols to overcome these limitations, thus opening up novel opportunities for Tα1 application. Herein, we summarize in a historical perspective the use of Tα1 in cancer, with particular reference to melanoma, hepatocellular carcinoma and lung cancer. We will discuss the current limitations of checkpoint inhibitors in clinical practice and the mechanisms at the basis of a potential application of Tα1 in combination protocols.
- Published
- 2019
- Full Text
- View/download PDF
38. Publisher Correction: Thymosin α1 represents a potential potent single-molecule-based therapy for cystic fibrosis.
- Author
-
Romani L, Oikonomou V, Moretti S, Iannitti RG, D'Adamo MC, Villella VR, Pariano M, Sforna L, Borghi M, Bellet MM, Fallarino F, Pallotta MT, Servillo G, Ferrari E, Puccetti P, Kroemer G, Pessia M, Maiuri L, Goldstein AL, and Garaci E
- Abstract
In the version of this article originally published, the amino acid sequence for Tα1 described in the Online Methods is incorrect. The sequence is described as "Ac-SDAAVDTSSEITTJDLKEKKEVVEEAEN-OH". It should be "Ac-SDAAVDTSSEITTKDLKEKKEVVEEAEN-OH". The error has been corrected in the HTML and PDF versions of this article.
- Published
- 2018
- Full Text
- View/download PDF
39. Author Correction: Thymosin α1 represents a potential potent single-molecule-based therapy for cystic fibrosis.
- Author
-
Romani L, Oikonomou V, Moretti S, Iannitti RG, D'Adamo MC, Villella VR, Pariano M, Sforna L, Borghi M, Bellet MM, Fallarino F, Pallotta MT, Servillo G, Ferrari E, Puccetti P, Kroemer G, Pessia M, Maiuri L, Goldstein AL, and Garaci E
- Abstract
In the version of this article originally published, some labels in Fig. 1f are incorrect. The "β-actin" labels on the second and fourth rows of blots should instead be "β-tubulin". The error has been corrected in the HTML and PDF versions of this article.
- Published
- 2018
- Full Text
- View/download PDF
40. Cellular proteostasis: a new twist in the action of thymosin α1.
- Author
-
Stincardini C, Renga G, Villella V, Pariano M, Oikonomou V, Borghi M, Bellet MM, Sforna L, Costantini C, Goldstein AL, Garaci E, and Romani L
- Subjects
- Animals, Cystic Fibrosis Transmembrane Conductance Regulator physiology, Humans, Immune Tolerance drug effects, Immune Tolerance genetics, Immunity, Innate drug effects, Immunity, Innate genetics, Inflammation genetics, Inflammation prevention & control, Signal Transduction drug effects, Thymalfasin pharmacology, Proteostasis drug effects, Proteostasis genetics, Thymalfasin physiology
- Abstract
Introduction: Thymosin alpha 1 (Tα1) is a naturally occurring polypeptide of 28 amino acids, whose mechanism of action is thought to be related to its ability to signal through innate immune receptors. Tα1 (ZADAXIN®) is used worldwide for treating viral infections, immunodeficiencies, and malignancies. Owing to its ability to activate the tolerogenic pathway of tryptophan catabolism - via the immunoregulatory enzyme indoleamine 2,3-dioxygenase - Tα1 potentiates immune tolerance mechanisms, breaking the vicious circle that perpetuates chronic inflammation in response to a variety of infectious noxae., Areas Covered: Tα1 has never been studied in Cystic fibrosis (CF) in which the hyperinflammatory state is associated with early and nonresolving activation of innate immunity, which impairs microbial clearance and promotes a self-sustaining condition of progressive lung damage. Optimal CF treatments should, indeed, not only rescue CF transmembrane conductance regulator protein localization and functionality but also alleviate the associated hyperinflammatory pathology. Because of the inherent complexity of the pathogenetic mechanisms, a multidrug approach is required., Expert Opinion: By providing a multipronged attack against CF, i.e. restraining inflammation and correcting the basic defect, Tα1 favorably opposed CF symptomatology in preclinical relevant disease settings, thus suggesting its possible exploitation for 'real-life' clinical efficacy in CF. This could represent a major conceptual advance in the CF field, namely the proposal of a drug with the unique activity to correct CFTR defects through regulation of inflammation.
- Published
- 2018
- Full Text
- View/download PDF
41. Reply to 'F508del-CFTR is not corrected by thymosin α1'.
- Author
-
Romani L, Stincardini C, Giovagnoli S, Paci M, Villella VR, Sforna L, Renga G, Bellet MM, Costantini C, Puccetti P, Kroemer G, Maiuri L, Pessia M, Goldstein A, and Garaci E
- Subjects
- Cell Line, Humans, Thymalfasin, Cystic Fibrosis, Cystic Fibrosis Transmembrane Conductance Regulator
- Published
- 2018
- Full Text
- View/download PDF
42. Thymosin α1 represents a potential potent single-molecule-based therapy for cystic fibrosis.
- Author
-
Romani L, Oikonomou V, Moretti S, Iannitti RG, D'Adamo MC, Villella VR, Pariano M, Sforna L, Borghi M, Bellet MM, Fallarino F, Pallotta MT, Servillo G, Ferrari E, Puccetti P, Kroemer G, Pessia M, Maiuri L, Goldstein AL, and Garaci E
- Subjects
- Animals, Autophagy drug effects, Blotting, Western, Cell Line, Chloride Channels drug effects, Chloride Channels metabolism, Cystic Fibrosis immunology, Cystic Fibrosis metabolism, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Cytokines immunology, Disease Models, Animal, Epithelial Cells metabolism, Fluorescent Antibody Technique, Humans, Immunohistochemistry, Immunoprecipitation, Indoleamine-Pyrrole 2,3,-Dioxygenase drug effects, Indoleamine-Pyrrole 2,3,-Dioxygenase immunology, Inflammation, Mice, Mice, Inbred CFTR, Patch-Clamp Techniques, Protein Stability drug effects, RAW 264.7 Cells, Respiratory Mucosa cytology, Thymalfasin, Thymosin pharmacology, Ubiquitin Thiolesterase drug effects, Ubiquitin Thiolesterase metabolism, Ubiquitination drug effects, Adjuvants, Immunologic pharmacology, Cystic Fibrosis genetics, Cystic Fibrosis Transmembrane Conductance Regulator drug effects, Cytokines drug effects, Epithelial Cells drug effects, Thymosin analogs & derivatives
- Abstract
Cystic fibrosis (CF) is caused by mutations in the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR) that compromise its chloride channel activity. The most common mutation, p.Phe508del, results in the production of a misfolded CFTR protein, which has residual channel activity but is prematurely degraded. Because of the inherent complexity of the pathogenetic mechanisms involved in CF, which include impaired chloride permeability and persistent lung inflammation, a multidrug approach is required for efficacious CF therapy. To date, no individual drug with pleiotropic beneficial effects is available for CF. Here we report on the ability of thymosin alpha 1 (Tα1)-a naturally occurring polypeptide with an excellent safety profile in the clinic when used as an adjuvant or an immunotherapeutic agent-to rectify the multiple tissue defects in mice with CF as well as in cells from subjects with the p.Phe508del mutation. Tα1 displayed two combined properties that favorably opposed CF symptomatology: it reduced inflammation and increased CFTR maturation, stability and activity. By virtue of this two-pronged action, Tα1 has strong potential to be an efficacious single-molecule-based therapeutic agent for CF.
- Published
- 2017
- Full Text
- View/download PDF
43. Histone Deacetylase SIRT1 Controls Proliferation, Circadian Rhythm, and Lipid Metabolism during Liver Regeneration in Mice.
- Author
-
Bellet MM, Masri S, Astarita G, Sassone-Corsi P, Della Fazia MA, and Servillo G
- Subjects
- Animals, Humans, Liver metabolism, Liver physiopathology, Liver Diseases genetics, Liver Diseases metabolism, Liver Diseases physiopathology, Mice, Mice, Knockout, Sirtuin 1 genetics, Cell Cycle, Cell Proliferation, Lipid Metabolism, Liver enzymology, Liver Diseases enzymology, Liver Regeneration, Sirtuin 1 metabolism
- Abstract
Liver regeneration offers a distinctive opportunity to study cell proliferation in vivo Mammalian silent information regulator 1 (SIRT1), a NAD
+ -dependent histone deacetylase, is an important regulator of various cellular processes, including proliferation, metabolism, and circadian rhythms. In the liver, SIRT1 coordinates the circadian oscillation of clock-controlled genes, including genes that encode enzymes involved in metabolic pathways. We performed partial hepatectomy in WT and liver-specific Sirt1-deficient mice and analyzed the expression of cell cycle regulators in liver samples taken at different times during the regenerative process, by real time PCR, Western blotting analysis, and immunohistochemistry. Lipidomic analysis was performed in the same samples by MS/HPLC. We showed that G1 /S progression was significantly affected by absence of SIRT1 in the liver, as well as circadian gene expression. This was associated to lipid accumulation due to defective fatty acid beta-oxidation. Our study revealed for the first time the importance of SIRT1 in the regulation of hepatocellular proliferation, circadian rhythms, and lipid metabolism during liver regeneration in mice. These results represent an additional step toward the characterization of SIRT1 function in the liver., (© 2016 by The American Society for Biochemistry and Molecular Biology, Inc.)- Published
- 2016
- Full Text
- View/download PDF
44. Impaired cell proliferation in regenerating liver of 3 β-hydroxysterol Δ14-reductase (TM7SF2) knock-out mice.
- Author
-
Bartoli D, Piobbico D, Bellet MM, Bennati AM, Roberti R, Della Fazia MA, and Servillo G
- Subjects
- Animals, Cell Proliferation, Endoplasmic Reticulum Stress, Fatty Liver complications, G1 Phase, Hepatectomy, Hepatocytes metabolism, Hepatocytes pathology, Lipids analysis, Male, Mice, Inbred C57BL, Mice, Knockout, Oxidoreductases deficiency, Oxidoreductases genetics, Receptors, Cytoplasmic and Nuclear genetics, Receptors, Cytoplasmic and Nuclear metabolism, S Phase, Lamin B Receptor, Liver Regeneration genetics, Oxidoreductases metabolism
- Abstract
The liver is the most important organ in cholesterol metabolism, which is instrumental in regulating cell proliferation and differentiation. The gene Tm7sf2 codifies for 3 β-hydroxysterol-Δ
14 -reductase (C14-SR), an endoplasmic reticulum resident protein catalyzing the reduction of C14-unsaturated sterols during cholesterol biosynthesis from lanosterol. In this study we analyzed the role of C14-SR in vivo during cell proliferation by evaluating liver regeneration in Tm7sf2 knockout (KO) and wild-type (WT) mice. Tm7sf2 KO mice showed no alteration in cholesterol content. However, accumulation and delayed catabolism of hepatic triglycerides was observed, resulting in persistent steatosis at all times post hepatectomy. Moreover, delayed cell cycle progression to the G1/S phase was observed in Tm7sf2 KO mice, resulting in reduced cell division at the time points examined. This was associated to abnormal ER stress response, leading to alteration in p53 content and, consequently, induction of p21 expression in Tm7sf2 KO mice. In conclusion, our results indicate that Tm7sf2 deficiency during liver regeneration alters lipid metabolism and generates a stress condition, which, in turn, transiently unbalances hepatocytes cell cycle progression.- Published
- 2016
- Full Text
- View/download PDF
45. Circadian clock proteins and immunity.
- Author
-
Curtis AM, Bellet MM, Sassone-Corsi P, and O'Neill LA
- Subjects
- Animals, Circadian Rhythm genetics, Circadian Rhythm immunology, Gene Expression Regulation, Humans, Nuclear Receptor Subfamily 1, Group D, Member 1 immunology, Circadian Rhythm physiology, Immunity, Nuclear Receptor Subfamily 1, Group D, Member 1 physiology
- Abstract
Immune parameters change with time of day and disruption of circadian rhythms has been linked to inflammatory pathologies. A circadian-clock-controlled immune system might allow an organism to anticipate daily changes in activity and feeding and the associated risk of infection or tissue damage to the host. Responses to bacteria have been shown to vary depending on time of infection, with mice being more at risk of sepsis when challenged ahead of their activity phase. Studies highlight the extent to which the molecular clock, most notably the core clock proteins BMAL1, CLOCK, and REV-ERBα, control fundamental aspects of the immune response. Examples include the BMAL1:CLOCK heterodimer regulating toll-like receptor 9 (TLR9) expression and repressing expression of the inflammatory monocyte chemokine ligand (CCL2) as well as REV-ERBα suppressing the induction of interleukin-6. Understanding the daily rhythm of the immune system could have implications for vaccinations and how we manage infectious and inflammatory diseases., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
46. Different functions of HOPS isoforms in the cell: HOPS shuttling isoform is determined by RIP cleavage system.
- Author
-
Castelli M, Piobbico D, Bartoli D, Pieroni S, Brunacci C, Bellet MM, Chiacchiaretta M, Della Fazia MA, and Servillo G
- Subjects
- Animals, Carrier Proteins genetics, Cell Line, Cell Nucleus metabolism, Chlorocebus aethiops, Computer Simulation, Cytosol metabolism, Humans, Intracellular Signaling Peptides and Proteins, Membrane Proteins, Mice, Nuclear Proteins genetics, Nucleophosmin, Organ Specificity, Protein Binding, Protein Conformation, Protein Isoforms genetics, Protein Isoforms metabolism, Proteolysis, Rats, Ubiquitin metabolism, Carrier Proteins metabolism, Nuclear Proteins metabolism
- Abstract
Hepatocyte odd protein shuttling (HOPS) moves between nucleus and cytoplasm. HOPS overexpression leads to cell cycle arrest in G 0/G 1, and HOPS knockdown causes centrosome alterations, with subsequent abnormal cell division. Recently, we demonstrated that HOPS acts as a functional bridge in NPM-p19(Arf) interactions. Here we show that HOPS is present in 3 different isoforms that play distinct intracellular functions. Although HOPS is a transmembrane ubiquitin, an isoform with intermediate molecular weight is cleaved from the membrane and released into the cytosol, to act as the shuttling protein. We identified a signal peptide peptidase structure in N-terminal membrane-bound HOPS that allows the regulated intramembrane proteolysis (RIP) system to control the relative amounts of the released, shuttling isoform capable of binding NPM. These results argue for distinct, isoform-specific functions of HOPS in the nucleolus, nucleus, and cytoplasm and provide insight into the dynamics of HOPS association with NPM, whose mutation and subsequent delocalization is found in 30% of acute myeloid leukemia patients.
- Published
- 2014
- Full Text
- View/download PDF
47. NEDD4 controls the expression of GUCD1, a protein upregulated in proliferating liver cells.
- Author
-
Bellet MM, Piobbico D, Bartoli D, Castelli M, Pieroni S, Brunacci C, Chiacchiaretta M, Del Sordo R, Fallarino F, Sidoni A, Puccetti P, Romani L, Servillo G, and Della Fazia MA
- Subjects
- Animals, Carcinoma, Hepatocellular metabolism, Carcinoma, Hepatocellular pathology, Cell Cycle, Cell Proliferation, Endosomal Sorting Complexes Required for Transport genetics, Guanylate Cyclase genetics, Humans, Liver cytology, Liver Neoplasms metabolism, Liver Neoplasms pathology, Mice, Nedd4 Ubiquitin Protein Ligases, Rats, Sprague-Dawley, Ubiquitin-Protein Ligases genetics, Up-Regulation, Endosomal Sorting Complexes Required for Transport metabolism, Guanylate Cyclase metabolism, Liver metabolism, Liver Regeneration genetics, Ubiquitin-Protein Ligases metabolism
- Abstract
Liver regeneration is a unique means of studying cell proliferation in vivo. Screening of a large cDNA library from regenerating liver has previously allowed us to identify and characterize a cluster of genes encoding proteins with important roles in proliferative processes. Here, by examining different rat and human tissues as well as cell lines, we characterized a highly conserved gene, guanylyl cyclase domain containing 1 (GUCD1), whose modulation occurs in liver regeneration and cell cycle progression in vitro. High-level expression of GUCD1 transcripts was observed in livers from patients with hepatocellular carcinoma. A yeast two-hybrid interaction assay, aimed at identifying any relevant interaction partners of GUCD1, revealed direct interactions with NEDD4-1 (E3 ubiquitin protein ligase neural precursor cell expressed, developmentally downregulated gene 4), resulting in control of GUCD1 stability. Thus, we have characterized expression and function of a ubiquitous protein, GUCD1, which might have a role in regulating normal and abnormal cell growth in the liver.
- Published
- 2014
- Full Text
- View/download PDF
48. Hepatocyte odd protein shuttling (HOPS) is a bridging protein in the nucleophosmin-p19 Arf network.
- Author
-
Castelli M, Pieroni S, Brunacci C, Piobbico D, Bartoli D, Bellet MM, Colombo E, Pelicci PG, Della Fazia MA, and Servillo G
- Subjects
- Animals, Carrier Proteins chemistry, Carrier Proteins genetics, Cell Cycle, Cell Line, Cyclin-Dependent Kinase Inhibitor p16 chemistry, Gene Knockout Techniques, Humans, Intracellular Signaling Peptides and Proteins, Membrane Proteins, Mice, Nuclear Proteins chemistry, Nuclear Proteins deficiency, Nuclear Proteins genetics, Nucleophosmin, Protein Multimerization, Protein Stability, Protein Structure, Quaternary, Protein Transport, Carrier Proteins metabolism, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Nuclear Proteins metabolism
- Abstract
Nucleophosmin (NPM), a ubiquitously and abundantly expressed protein, occurs in the nucleolus, shuttling between the nucleoplasm and cytoplasm. The NPM gene is mutated in almost 30% of human acute myeloid leukemia cells. NPM interacts with p53 and p19(Arf), directs localization of p19(Arf) in the nucleolus and protects the latter from degradation. Hepatocyte odd protein shuttling (HOPS) is also a ubiquitously expressed protein that moves between the nucleus and cytoplasm. Within the nucleus of resting cells, HOPS overexpression causes cell cycle arrest in G0/G1. HOPS knockdown causes centrosome hyperamplification leading to multinucleated cells and the formation of micronuclei. We demonstrate a direct interaction of HOPS with NPM and p19(Arf), resulting in a functionally active trimeric complex. NPM appeared to regulate HOPS half-life, which, in turn, stabilized p19(Arf) and controlled its localization in the nucleolus. These findings suggest that HOPS acts as a functional bridge in the interaction between NPM and p19(Arf), providing new mechanistic insight into how NPM and p19(Arf) will oppose tumor cell proliferation.
- Published
- 2013
- Full Text
- View/download PDF
49. Circadian clock regulates the host response to Salmonella.
- Author
-
Bellet MM, Deriu E, Liu JZ, Grimaldi B, Blaschitz C, Zeller M, Edwards RA, Sahar S, Dandekar S, Baldi P, George MD, Raffatellu M, and Sassone-Corsi P
- Subjects
- Animals, CLOCK Proteins deficiency, CLOCK Proteins genetics, CLOCK Proteins immunology, Cecum immunology, Cecum metabolism, Cecum microbiology, Cells, Cultured, Circadian Clocks genetics, Cluster Analysis, Cytokines genetics, Cytokines metabolism, Gene Expression Profiling, Gene Regulatory Networks genetics, Gene Regulatory Networks immunology, Host-Pathogen Interactions immunology, Inflammation Mediators immunology, Inflammation Mediators metabolism, Lipopolysaccharides immunology, Macrophages immunology, Macrophages metabolism, Macrophages microbiology, Mice, Mice, Inbred C57BL, Mice, Knockout, Oligonucleotide Array Sequence Analysis, Reverse Transcriptase Polymerase Chain Reaction, Salmonella Infections, Animal genetics, Salmonella Infections, Animal microbiology, Salmonella typhimurium physiology, Time Factors, Circadian Clocks immunology, Cytokines immunology, Salmonella Infections, Animal immunology, Salmonella typhimurium immunology
- Abstract
Organisms adapt to day-night cycles through highly specialized circadian machinery, whose molecular components anticipate and drive changes in organism behavior and metabolism. Although many effectors of the immune system are known to follow daily oscillations, the role of the circadian clock in the immune response to acute infections is not understood. Here we show that the circadian clock modulates the inflammatory response during acute infection with the pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium). Mice infected with S. Typhimurium were colonized to higher levels and developed a higher proinflammatory response during the early rest period for mice, compared with other times of the day. We also demonstrate that a functional clock is required for optimal S. Typhimurium colonization and maximal induction of several proinflammatory genes. These findings point to a clock-regulated mechanism of activation of the immune response against an enteric pathogen and may suggest potential therapeutic strategies for chronopharmacologic interventions.
- Published
- 2013
- Full Text
- View/download PDF
50. Pharmacological modulation of circadian rhythms by synthetic activators of the deacetylase SIRT1.
- Author
-
Bellet MM, Nakahata Y, Boudjelal M, Watts E, Mossakowska DE, Edwards KA, Cervantes M, Astarita G, Loh C, Ellis JL, Vlasuk GP, and Sassone-Corsi P
- Subjects
- ARNTL Transcription Factors metabolism, Animals, CLOCK Proteins genetics, CLOCK Proteins metabolism, Cell Line, Chromatin metabolism, Circadian Rhythm drug effects, Gene Expression Regulation drug effects, Heterocyclic Compounds, 4 or More Rings pharmacology, Humans, Liver drug effects, Liver metabolism, Mice, Mice, Knockout, NAD metabolism, Transcription, Genetic drug effects, Circadian Rhythm genetics, Enzyme Activators pharmacology, Sirtuin 1 metabolism
- Abstract
Circadian rhythms govern a wide variety of physiological and metabolic functions in many organisms, from prokaryotes to humans. We previously reported that silent information regulator 1 (SIRT1), a NAD(+)-dependent deacetylase, contributes to circadian control. In addition, SIRT1 activity is regulated in a cyclic manner in virtue of the circadian oscillation of the coenzyme NAD(+). Here we used specific SIRT1 activator compounds both in vitro and in vivo. We tested a variety of compounds to show that the activation of SIRT1 alters CLOCK:BMAL1-driven transcription in different systems. Activation of SIRT1 induces repression of circadian gene expression and decreases H3 K9/K14 acetylation at corresponding promoters in a time-specific manner. Specific activation of SIRT1 was demonstrated in vivo using liver-specific SIRT1-deficient mice, where the effect of SIRT1 activator compounds was shown to be dependent on SIRT1. Our findings demonstrate that SIRT1 can fine-tune circadian rhythm and pave the way to the development of pharmacological strategies to address a broad range of therapeutic indications.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.