46 results on '"F. Michielin"'
Search Results
2. 745P Clinical activity, safety, and PK/PD from the first in human study (NP41300) of RO7247669, a PD1-LAG3 bispecific antibody
- Author
-
K.S. Rohrberg, E. Garralda, E. Calvo, V. Moreno Garcia, M. Guidi, D.G. Kraus, C. McIntyre, H. Kao, L. Codarri Deak, F. Michielin, T. Liu, M. Muecke, C. Markert, and I. Melero
- Subjects
Oncology ,Hematology - Published
- 2022
- Full Text
- View/download PDF
3. The emergence of the circadian clock network in hiPSC-derived hepatocytes on chip
- Author
-
Gagliano O, S. Cascione, F. Michielin, and N. Elvassore
- Subjects
Circadian clock ,Development ,Hepatic differentiation ,Hepatocytes ,Human pluripotent stem cell ,ARNTL Transcription Factors ,CLOCK Proteins ,Circadian Rhythm ,Female ,Humans ,Liver ,Pregnancy ,Circadian Clocks ,Induced Pluripotent Stem Cells ,Biophysics ,Cell Biology ,Biochemistry ,Molecular Biology - Abstract
The circadian clock has paramount implications in physiology and pathology. Although the circadian clock has been widely investigated in adults, up to now very little is known about how circadian rhythms emerge during embryonic development. Some studies about the ontology of the circadian system are focused on the development of the central pacemaker, whereas there is still no agreement about the development of the circadian clock in peripheral tissues. Our work represents the first attempt at investigating the onset of peripheral circadian clocks in the liver, which has a central role in controlling several aspects of human physiology. We profile the emergence of the circadian genes during the transition from the initial state of human pluripotency to the final state of hepatic maturation. We demonstrate that circadian rhythmicity is absent in human pluripotent stem cells, and it arises gradually during the process of hepatic commitment. The clock genes expression reaches a peak at the hepatic progenitor stage. At this point o hiPSC-derived f differentiation the gene oscillations start to be observed with a period of 13 h and approaches 24 h in a later stage when the clock primary feedback loop starts working properly. At the end of differentiation, circadian rhythmicity appears, with genes of primary and secondary feedback loops in antiphase (CLOCK, BMAL1 and REV-ERBα) a sign that the system becomes to be functional.
- Published
- 2022
4. Development of a microfluidic approach for the real-time analysis of extrinsic TGF-β signalling
- Author
-
F, Michielin, M, Vetralla, C, Bolego, O, Gagliano, M, Montagner, and N, Elvassore
- Published
- 2020
- Full Text
- View/download PDF
5. [Epidemiology and prevention of rheumatic fever in Rio Grande do Sul]
- Author
-
F, Michielin, A A, Pretto, J C, Prataviera, J L, Holz, R, Paternoster, M, Catalunha, F, Michielin Filho, and J H, Michielin
- Subjects
Male ,Risk Factors ,Humans ,Rheumatic Fever ,Brazil - Published
- 1994
6. Author Correction: Single-cell guided prenatal derivation of primary fetal epithelial organoids from human amniotic and tracheal fluids.
- Author
-
Gerli MFM, Calà G, Beesley MA, Sina B, Tullie L, Sun KY, Panariello F, Michielin F, Davidson JR, Russo FM, Jones BC, Lee DDH, Savvidis S, Xenakis T, Simcock IC, Straatman-Iwanowska AA, Hirst RA, David AL, O'Callaghan C, Olivo A, Eaton S, Loukogeorgakis SP, Cacchiarelli D, Deprest J, Li VSW, Giobbe GG, and De Coppi P
- Published
- 2025
- Full Text
- View/download PDF
7. Sepsis death risk factor score based on systemic inflammatory response syndrome, quick sequential organ failure assessment, and comorbidities.
- Author
-
Orsatti VN, Ribeiro VST, de Oliveira Montenegro C, Costa CJ, Raboni EA, Sampaio ER, Michielin F, Gasparetto J, Telles JP, and Tuon FF
- Subjects
- Aged, Female, Humans, Male, Middle Aged, Brazil epidemiology, Retrospective Studies, Risk Assessment methods, Risk Factors, Comorbidity, Hospital Mortality, Organ Dysfunction Scores, Sepsis mortality, Systemic Inflammatory Response Syndrome mortality, Systemic Inflammatory Response Syndrome epidemiology
- Abstract
Objective: In this study, we aimed to evaluate the death risk factors of patients included in the sepsis protocol bundle, using clinical data from qSOFA, SIRS, and comorbidities, as well as development of a mortality risk score., Design: This retrospective cohort study was conducted between 2016 and 2021., Setting: Two university hospitals in Brazil., Participants: Patients with sepsis., Interventions: Several clinical and laboratory data were collected focused on SIRS, qSOFA, and comorbidities., Main Variable of Interest: In-hospital mortality was the primary outcome variable. A mortality risk score was developed after logistic regression analysis., Results: A total of 1,808 patients were included with a death rate of 36%. Ten variables remained independent factors related to death in multivariate analysis: temperature ≥38 °C (odds ratio [OR] = 0.65), previous sepsis (OR = 1.42), qSOFA ≥ 2 (OR = 1.43), leukocytes >12,000 or <4,000 cells/mm
3 (OR = 1.61), encephalic vascular accident (OR = 1.88), age >60 years (OR = 1.93), cancer (OR = 2.2), length of hospital stay before sepsis >7 days (OR = 2.22,), dialysis (OR = 2.51), and cirrhosis (OR = 3.97). Considering the equation of the binary regression logistic analysis, the score presented an area under curve of 0.668, is not a potential model for death prediction., Conclusions: Several risk factors are independently associated with mortality, allowing the development of a prediction score based on qSOFA, SIRS, and comorbidities data, however, the performance of this score is low., (Copyright © 2024 Elsevier España, S.L.U. and SEMICYUC. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
8. Single-cell guided prenatal derivation of primary fetal epithelial organoids from human amniotic and tracheal fluids.
- Author
-
Gerli MFM, Calà G, Beesley MA, Sina B, Tullie L, Sun KY, Panariello F, Michielin F, Davidson JR, Russo FM, Jones BC, Lee DDH, Savvidis S, Xenakis T, Simcock IC, Straatman-Iwanowska AA, Hirst RA, David AL, O'Callaghan C, Olivo A, Eaton S, Loukogeorgakis SP, Cacchiarelli D, Deprest J, Li VSW, Giobbe GG, and De Coppi P
- Subjects
- Pregnancy, Female, Humans, Amniotic Fluid metabolism, Prenatal Care, Lung metabolism, Organoids metabolism, Hernias, Diaphragmatic, Congenital metabolism
- Abstract
Isolation of tissue-specific fetal stem cells and derivation of primary organoids is limited to samples obtained from termination of pregnancies, hampering prenatal investigation of fetal development and congenital diseases. Therefore, new patient-specific in vitro models are needed. To this aim, isolation and expansion of fetal stem cells during pregnancy, without the need for tissue samples or reprogramming, would be advantageous. Amniotic fluid (AF) is a source of cells from multiple developing organs. Using single-cell analysis, we characterized the cellular identities present in human AF. We identified and isolated viable epithelial stem/progenitor cells of fetal gastrointestinal, renal and pulmonary origin. Upon culture, these cells formed clonal epithelial organoids, manifesting small intestine, kidney tubule and lung identity. AF organoids exhibit transcriptomic, protein expression and functional features of their tissue of origin. With relevance for prenatal disease modeling, we derived lung organoids from AF and tracheal fluid cells of congenital diaphragmatic hernia fetuses, recapitulating some features of the disease. AF organoids are derived in a timeline compatible with prenatal intervention, potentially allowing investigation of therapeutic tools and regenerative medicine strategies personalized to the fetus at clinically relevant developmental stages., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
9. Atezolizumab plus Magrolimab, Niraparib, or Tocilizumab versus Atezolizumab Monotherapy in Platinum-Refractory Metastatic Urothelial Carcinoma: A Phase Ib/II Open-Label, Multicenter, Randomized Umbrella Study (MORPHEUS Urothelial Carcinoma).
- Author
-
Drakaki A, Powles T, Bamias A, Martin-Liberal J, Shin SJ, Friedlander T, Tosi D, Park C, Gomez-Roca C, Joly Lobbedez F, Castellano D, Morales-Barrera R, Moreno-Candilejo I, Fléchon A, Yuen K, Rishipathak D, DuPree K, Young F, Michielin F, Shemesh CS, Steinberg EE, Williams P, and Lee JL
- Subjects
- Humans, Antineoplastic Combined Chemotherapy Protocols adverse effects, Platinum therapeutic use, Carcinoma, Transitional Cell pathology, Urinary Bladder Neoplasms, Urologic Neoplasms pathology
- Abstract
Purpose: The MORPHEUS platform was designed to identify early efficacy signals and evaluate the safety of novel immunotherapy combinations across cancer types. The phase Ib/II MORPHEUS-UC trial (NCT03869190) is evaluating atezolizumab plus magrolimab, niraparib, or tocilizumab in platinum-refractory locally advanced or metastatic urothelial carcinoma (mUC). Additional treatment combinations were evaluated and will be reported separately., Patients and Methods: Patients had locally advanced or mUC that progressed during or following treatment with a platinum-containing regimen. The primary efficacy endpoint was investigator-assessed objective response rate (ORR). Key secondary endpoints included investigator-assessed progression-free survival (PFS) and overall survival (OS). Safety and exploratory biomarker analyses were also conducted., Results: Seventy-six patients were randomized to receive either atezolizumab plus magrolimab (n = 16), atezolizumab plus niraparib (n = 15), atezolizumab plus tocilizumab (n = 15), or atezolizumab monotherapy (control; n = 30). No additive benefit in ORR, PFS, or OS was seen in the treatment arms versus the control. The best confirmed ORR was 26.7% with atezolizumab plus magrolimab, 6.7% with atezolizumab plus niraparib, 20.0% with atezolizumab plus tocilizumab, and 27.6% with atezolizumab monotherapy. Overall, the treatment combinations were tolerable, and adverse events were consistent with each agent's known safety profile. Trends were observed for shrinkage of programmed death-ligand 1-positive tumors (atezolizumab, atezolizumab plus magrolimab, atezolizumab plus tocilizumab), inflamed tumors, or tumors with high mutational burden (atezolizumab), and immune excluded tumors (atezolizumab plus magrolimab)., Conclusions: The evaluated regimens in MORPHEUS-UC were tolerable. However, response rates for the combinations did not meet the criteria for further development in platinum-experienced locally advanced or mUC., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
10. Lung viral infection modelling in a bioengineered whole-organ.
- Author
-
Tommasini F, Benoist T, Shibuya S, Woodall MNJ, Naldi E, Palor M, Orr JC, Giobbe GG, Maughan EF, Saleh T, Gjinovci A, Hutchinson JC, Arthurs OJ, Janes SM, Elvassore N, Hynds RE, Smith CM, Michielin F, Pellegata AF, and De Coppi P
- Subjects
- Rats, Humans, Animals, Lung, Epithelial Cells, Tissue Scaffolds chemistry, COVID-19, Pneumonia, Virus Diseases
- Abstract
Lung infections are one of the leading causes of death worldwide, and this situation has been exacerbated by the emergence of COVID-19. Pre-clinical modelling of viral infections has relied on cell cultures that lack 3D structure and the context of lung extracellular matrices. Here, we propose a bioreactor-based, whole-organ lung model of viral infection. The bioreactor takes advantage of an automated system to achieve efficient decellularization of a whole rat lung, and recellularization of the scaffold using primary human bronchial cells. Automatization allowed for the dynamic culture of airway epithelial cells in a breathing-mimicking setup that led to an even distribution of lung epithelial cells throughout the distal regions. In the sealed bioreactor system, we demonstrate proof-of-concept for viral infection within the epithelialized lung by infecting primary human airway epithelial cells and subsequently injecting neutrophils. Moreover, to assess the possibility of drug screening in this model, we demonstrate the efficacy of the broad-spectrum antiviral remdesivir. This whole-organ scale lung infection model represents a step towards modelling viral infection of human cells in a 3D context, providing a powerful tool to investigate the mechanisms of the early stages of pathogenic infections and the development of effective treatment strategies for respiratory diseases., 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. Published by Elsevier Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
11. Hydrogel-in-hydrogel live bioprinting for guidance and control of organoids and organotypic cultures.
- Author
-
Urciuolo A, Giobbe GG, Dong Y, Michielin F, Brandolino L, Magnussen M, Gagliano O, Selmin G, Scattolini V, Raffa P, Caccin P, Shibuya S, Scaglioni D, Wang X, Qu J, Nikolic M, Montagner M, Galea GL, Clevers H, Giomo M, De Coppi P, and Elvassore N
- Subjects
- Hydrogels chemistry, Tissue Engineering methods, Cell Polarity, Lung, Bioprinting, Organoids
- Abstract
Three-dimensional hydrogel-based organ-like cultures can be applied to study development, regeneration, and disease in vitro. However, the control of engineered hydrogel composition, mechanical properties and geometrical constraints tends to be restricted to the initial time of fabrication. Modulation of hydrogel characteristics over time and according to culture evolution is often not possible. Here, we overcome these limitations by developing a hydrogel-in-hydrogel live bioprinting approach that enables the dynamic fabrication of instructive hydrogel elements within pre-existing hydrogel-based organ-like cultures. This can be achieved by crosslinking photosensitive hydrogels via two-photon absorption at any time during culture. We show that instructive hydrogels guide neural axon directionality in growing organotypic spinal cords, and that hydrogel geometry and mechanical properties control differential cell migration in developing cancer organoids. Finally, we show that hydrogel constraints promote cell polarity in liver organoids, guide small intestinal organoid morphogenesis and control lung tip bifurcation according to the hydrogel composition and shape., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
12. Human Pluripotent Stem Cell-Derived Micropatterned Ectoderm Allows Cell Sorting of Meso-Endoderm Lineages.
- Author
-
Yang Y, Laterza C, Stuart HT, Michielin F, Gagliano O, Urciuolo A, and Elvassore N
- Abstract
The human developmental processes during the early post-implantation stage instruct the specification and organization of the lineage progenitors into a body plan. These processes, which include patterning, cell sorting, and establishment of the three germ layers, have been classically studied in non-human model organisms and only recently, through micropatterning technology, in a human-specific context. Micropatterning technology has unveiled mechanisms during patterning and germ layer specification; however, cell sorting and their segregation in specific germ layer combinations have not been investigated yet in a human-specific in vitro system. Here, we developed an in vitro model of human ectodermal patterning, in which human pluripotent stem cells (hPSCs) self-organize to form a radially regionalized neural and non-central nervous system (CNS) ectoderm. We showed that by using micropatterning technology and by modulating BMP and WNT signals, we can regulate the appearance and spatial distribution of the different ectodermal populations. This pre-patterned ectoderm can be used to investigate the cell sorting behavior of hPSC-derived meso-endoderm cells, with an endoderm that segregates from the neural ectoderm. Thus, the combination of micro-technology with germ layer cross-mixing enables the study of cell sorting of different germ layers in a human context., 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., (Copyright © 2022 Yang, Laterza, Stuart, Michielin, Gagliano, Urciuolo and Elvassore.)
- Published
- 2022
- Full Text
- View/download PDF
13. Anti-CSF-1R emactuzumab in combination with anti-PD-L1 atezolizumab in advanced solid tumor patients naïve or experienced for immune checkpoint blockade.
- Author
-
Gomez-Roca C, Cassier P, Zamarin D, Machiels JP, Perez Gracia JL, Stephen Hodi F, Taus A, Martinez Garcia M, Boni V, Eder JP, Hafez N, Sullivan R, Mcdermott D, Champiat S, Aspeslagh S, Terret C, Jegg AM, Jacob W, Cannarile MA, Ries C, Korski K, Michielin F, Christen R, Babitzki G, Watson C, Meneses-Lorente G, Weisser M, Rüttinger D, Delord JP, and Marabelle A
- Subjects
- Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal, Humanized, Fatigue chemically induced, Humans, Immune Checkpoint Inhibitors, Ligands, Receptor Protein-Tyrosine Kinases, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung pathology, Lung Neoplasms drug therapy, Melanoma drug therapy, Urinary Bladder Neoplasms drug therapy
- Abstract
Background: This phase 1b study (NCT02323191) evaluated the safety, antitumor activity, pharmacokinetics, and pharmacodynamics of colony-stimulating factor-1 receptor-blocking monoclonal antibody (mAb) emactuzumab in combination with the programmed cell death-1 ligand (PD-L1)-blocking mAb atezolizumab in patients with advanced solid tumors naïve or experienced for immune checkpoint blockers (ICBs)., Methods: Emactuzumab (500-1350 mg flat) and atezolizumab (1200 mg flat) were administered intravenously every 3 weeks. Dose escalation of emactuzumab was conducted using the 3+3 design up to the maximum tolerated dose (MTD) or optimal biological dose (OBD). Extension cohorts to evaluate pharmacodynamics and clinical activity were conducted in metastatic ICB-naive urothelial bladder cancer (UBC) and ICB-pretreated melanoma (MEL), non-small cell lung cancer (NSCLC) and UBC patients., Results: Overall, 221 patients were treated. No MTD was reached and the OBD was determined at 1000 mg of emactuzumab in combination with 1200 mg of atezolizumab. Grade ≥3 treatment-related adverse events occurred in 25 (11.3%) patients of which fatigue and rash were the most common (14 patients (6.3%) each). The confirmed objective response rate (ORR) was 9.8% for ICB-naïve UBC, 12.5% for ICB-experienced NSCLC, 8.3% for ICB-experienced UBC and 5.6% for ICB-experienced MEL patients, respectively. Tumor biopsy analyses demonstrated increased activated CD8 +tumor infiltrating T lymphocytes (TILs) associated with clinical benefit in ICB-naïve UBC patients and less tumor-associated macrophage (TAM) reduction in ICB-experienced compared with ICB-naïve patients., Conclusion: Emactuzumab in combination with atezolizumab demonstrated a manageable safety profile with increased fatigue and skin rash over usual atezolizumab monotherapy. A considerable ORR was particularly seen in ICB-experienced NSCLC patients. Increase ofCD8 +TILs under therapy appeared to be associated with persistence of a TAM subpopulation., Competing Interests: Competing interests: CG-R: Invited Speaker: BMS, Eisai, Pierre Fabre, Roche/Genentech; Coordinating PI: BMS; Steering Committee Member: BMS; Local PI: Foundation Medicine; Steering Committee Member: Genentech; Research Grant: Roche/Genentech; AM: Stock ownership Pegascy, Hifibio Therapeutics, Shattuck Labs, Centessa Pharmaceuticals; Honoraria: BMS, AstraZeneca/MedImmune, Oncovir; Consulting and advisory activities: Lytix Biopharma, Eisai, Pierre Fabre, AstraZeneca, Servier, Roche, Redx Pharma, Sotio, Innate Pharma, ImCheck Therapeutics, MSD, OSE Immunotherapeutics, HIFIBIO Therapeutics, MedinCell, Centessa Pharmaceuticals; Speaker’s bureau: BMS; Research funding: BMS, Boehringer Ingelheim, Transgene, MSD; Travel expenses: MSD, AstraZeneca; SC: Honoraria: Amgen, AstraZeneca, BMS, EISAI, Janssen, MSD, Novartis and Roche; Principal Investigator of Clinical Trials for: Amgen, MSD, Sanofi Aventis, Transgene; Advisory Board: Alderaan Biotechnology, Amgen, AstraZeneca, Oncovita, Seagen, Ultrahuman; Travel and congress: AstraZeneca, MSD, Roche; Principal/sub-investigator of clinical trials for: Abbvie, Adaptimmune, Adlai Nortye USA Inc, Aduro Biotech, Agios Pharmaceuticals, Amgen, Argen-X Bvba, Astex Pharmaceuticals, Astra Zeneca Ab, Aveo, Basilea Pharmaceutica International Ltd, Bayer Healthcare Ag, Bbb Technologies Bv, Beigene, BicycleTx Ltd, Blueprint Medicines, Boehringer Ingelheim, Boston Pharmaceuticals, Bristol Myers Squibb, Ca, Celgene Corporation, Chugai Pharmaceutical Co, Clovis Oncology, Cullinan-Apollo, Curevac, Daiichi Sankyo, Debiopharm, Eisai, Eisai Limited, Eli Lilly, Exelixis, Faron Pharmaceuticals Ltd, Forma Tharapeutics, Gamamabs, Genentech, Glaxosmithkline, H3 Biomedicine, Hoffmann La Roche Ag, Imcheck Therapeutics, Innate Pharma, Institut De Recherche Pierre Fabre, Iris Servier, Iteos Belgium SA, Janssen Cilag, Janssen Research Foundation, Kura Oncology, Kyowa Kirin Pharm. Dev, Lilly France, Loxo Oncology, Lytix Biopharma As, Medimmune, Menarini Ricerche, Merck Sharp & Dohme Chibret, Merrimack Pharmaceuticals, Merus, Millennium Pharmaceuticals, Molecular Partners Ag, Nanobiotix, Nektar Therapeutics, Novartis Pharma, Octimet Oncology Nv, Oncoethix, Oncopeptides, Orion Pharma, Ose Pharma, Pfizer, Pharma Mar, Pierre Fabre, Medicament, Roche, Sanofi Aventis, Seattle Genetics, Sotio A.S, Syros Pharmaceuticals, Taiho Pharma, Tesaro, Turning Point Therapeutics, Xencor; Research Grants from: Astrazeneca, BMS, Boehringer Ingelheim, GSK, INCA, Janssen Cilag, Merck, Novartis, Pfizer, Roche, SanofiNon-financial support (drug supplied) from Astrazeneca, Bayer, BMS, Boringher Ingelheim, GSK, Medimmune, Merck, NH TherAGuiX, Pfizer, Roche; SA: Speakers bureau: Pfizer, Roche, Sanofi and BMSAdvisory board: Sanofi; PC: Honoraria: Novartis, Roche/Genentech, Amgen, Astra Zeneca, Merck Serono; Research Funding: Novartis, Roche/Genentech, Lilly, lueprint Medicines, Bayer, Astra Zeneca, Celgene, Plexxikon, Abbvie, BMS, Merck Serono, Merck Sharp and Dohme, Taiho Pharmaceutical, Toray Industries, Transgene, Loxo, GSK, Innate Pharma, Janssen; Travel expenses: Roche, Amgen, Novartis, BMS, MSD, Netris Pharma, Bayer, Merck Serono; DZ: Reports research support from: Roche, Astra Zeneca, and Plexxikon; Personal/consultancy fees from Synlogic Therapeutics, GSK, Roche, Xencor, Memgen, Immunos, Celldex, Calidi, and Agenus; J-PM: Advisory board member or speaker with honoraria: Pfizer, Roche, Astra/Zeneca, Bayer, Innate, Merck Serono, Boerhinger, BMS, Novartis, Janssen, Incyte, Cue Biopharma, ALX Oncology, iTEOS, eTheRNATravel expenses: Amgen, BMS, Pfizer, MSDData safety monitoring board with honoraria: Debio, Nanobiotix, Psioxus; Uncompensated advisory role: MSD; JLPG; Research grants and support: Roche, BMS, MSD, Seattle Genetics. Speakers bureau and advisory boards: Roche, BMS, Ipsen, MSD, Seattle Genetics. Travel support: Roche, MSD, BMS; FSH: Consulting: BMS, Merck, EMD Serono, Novartis, Sanofi, Psioxus Therapeutics, Pieris Pharmacutical, Corner Therapeutics, Eisai, Idera, Takeda, Genentech/Roche; Advisory Board: Compass Therapeutics, Apricity Scientific, Pionyr, Torque, Rheos, Bicara, Checkpoint Therapeutics, Bioentre, Gossamer, Iovance; ATG: Personal fees from: Boehringer-Ingelheim, BMS, MSD, Roche, Pfizer, Astra Zeneca, Tesaro-GSK and non-financial support from Boehringer-Ingelheim, Lilly and RocheMaria Martinez Garcia; Research grants and support: Roche, BMS, MSD, Seattle Genetics; Speakers bureau and advisory boards: Roche, BMS, Ipsen, MSD, Seattle Genetics; Travel support: Roche, MSD, BMS; VB: Consulting or Advisory Role: Puma Biotechnology; Ideaya Biosciences; Loxo Therapeutics, CytomX Therapeutics; Guidepoint; Oncoart; Amunix; Institutional financial support for clinical trials from: Abbvie, ACEO, Adaptaimmune, Amcure, AMGEN, AstraZeneca, BMS, Cytomx, GSK, Genentech/Roche, H3, Incyte, Janssen, Kura, Lilly, Loxo, Nektar, Macrogenics, Menarini, Merck, Merus, Nanobiotix, Novartis, Pfizer, PharmaMar, Principia, PUMA, Sanofi, Taiho, Tesaro, BeiGene, Transgene, Takeda, Incyte, Innovio, MSD, PsiOxus, Seattle Genetics, Mersana, GSK, Daiichi, Nektar, Astellas, ORCA, Boston Therapeutics, Dynavax, DebioPharm, Boehringen Ingelheim, Regeneron, Millenium, Synthon, Spectrum, Rigontec, Zenith; JPE: The author declares no potential conflicts of interest. NH: The author declares no potential conflicts of interest. RS: Consultant/advisory boards: Asana Biosciences, AstraZeneca, Bristol-Myers Squibb, Eisai, Iovance, Merck, Novartis, OncoSec, Pfizer, Replimune; Research funding: Amgen, Merck; DM: Consulting and honoraria: BMS, Pfizer, Merck, Alkermes Inc., EMD Serono, Eli Lilly and Company, Iovance, Eisai Inc., Werewolf Therapeutics, Calithera Biosciences; Research support: BMS, Merck, Genentech, Pfizer, Exelixis, X4 Pharma, Alkermes Inc; MAC: Sponsor employee and sponsor stock ownership; A-MJ: Former sponsor employee and has patent issued in the use of emactuzumab; WJ: Sponsor employee and sponsor stock ownership; CR: Former Roche employee and has patent issued in the use of emactuzumab. Consultant for Verseau Therapeutics, Ridgeline Discovery, iOmx Therapeutics AG; KK: Sponsor employee and Roche stocks; GB: Sponsor employee; FM: Sponsor employee; RC: Sponsor employee and Roche stocks; CW: Sponsor consultantGeorgina Meneses-LorenteSponsor employee; MW: Sponsor employee, stock options, and has patent issued in the use of emactuzumab; DR: Sponsor employee, sponsor stock ownership and has patent issued in the use of emactuzumab; J-PD: Consulting/Advisory: Novartis, Roche/Genentech, BMS, MSD; Research funding: Genentech, BMS, MSD, Astra Zeneca, Transgene; CT: Research funding GSK, travel expenses Mundipharma., (© 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
14. The emergence of the circadian clock network in hiPSC-derived hepatocytes on chip.
- Author
-
O G, Cascione S, Michielin F, and Elvassore N
- Subjects
- ARNTL Transcription Factors genetics, ARNTL Transcription Factors metabolism, CLOCK Proteins genetics, CLOCK Proteins metabolism, Circadian Rhythm genetics, Female, Hepatocytes metabolism, Humans, Liver metabolism, Pregnancy, Circadian Clocks genetics, Induced Pluripotent Stem Cells metabolism
- Abstract
The circadian clock has paramount implications in physiology and pathology. Although the circadian clock has been widely investigated in adults, up to now very little is known about how circadian rhythms emerge during embryonic development. Some studies about the ontology of the circadian system are focused on the development of the central pacemaker, whereas there is still no agreement about the development of the circadian clock in peripheral tissues. Our work represents the first attempt at investigating the onset of peripheral circadian clocks in the liver, which has a central role in controlling several aspects of human physiology. We profile the emergence of the circadian genes during the transition from the initial state of human pluripotency to the final state of hepatic maturation. We demonstrate that circadian rhythmicity is absent in human pluripotent stem cells, and it arises gradually during the process of hepatic commitment. The clock genes expression reaches a peak at the hepatic progenitor stage. At this point o hiPSC-derived f differentiation the gene oscillations start to be observed with a period of 13 h and approaches 24 h in a later stage when the clock primary feedback loop starts working properly. At the end of differentiation, circadian rhythmicity appears, with genes of primary and secondary feedback loops in antiphase (CLOCK, BMAL1 and REV-ERBα) a sign that the system becomes to be functional., 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 © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
15. Mitochondrial fission links ECM mechanotransduction to metabolic redox homeostasis and metastatic chemotherapy resistance.
- Author
-
Romani P, Nirchio N, Arboit M, Barbieri V, Tosi A, Michielin F, Shibuya S, Benoist T, Wu D, Hindmarch CCT, Giomo M, Urciuolo A, Giamogante F, Roveri A, Chakravarty P, Montagner M, Calì T, Elvassore N, Archer SL, De Coppi P, Rosato A, Martello G, and Dupont S
- Subjects
- Actin-Related Protein 2-3 Complex metabolism, Actins metabolism, Animals, Breast Neoplasms genetics, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Line, Transformed, Cell Line, Tumor, Cell-Matrix Junctions drug effects, Cell-Matrix Junctions metabolism, Cell-Matrix Junctions pathology, Dynamins metabolism, Extracellular Matrix genetics, Extracellular Matrix metabolism, Extracellular Matrix pathology, Female, Gene Expression Regulation, Neoplastic, Humans, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lung Neoplasms secondary, Mice, Inbred BALB C, Microfilament Proteins metabolism, Mitochondria genetics, Mitochondria metabolism, Mitochondria pathology, Mitochondrial Proteins metabolism, NF-E2-Related Factor 2 metabolism, Nuclear Proteins metabolism, Oxidation-Reduction, Oxidative Stress, Peptide Elongation Factors metabolism, Tumor Microenvironment, Mice, Antineoplastic Agents pharmacology, Breast Neoplasms drug therapy, Drug Resistance, Neoplasm, Energy Metabolism drug effects, Extracellular Matrix drug effects, Lung Neoplasms drug therapy, Mechanotransduction, Cellular drug effects, Mitochondria drug effects, Mitochondrial Dynamics drug effects
- Abstract
Metastatic breast cancer cells disseminate to organs with a soft microenvironment. Whether and how the mechanical properties of the local tissue influence their response to treatment remains unclear. Here we found that a soft extracellular matrix empowers redox homeostasis. Cells cultured on a soft extracellular matrix display increased peri-mitochondrial F-actin, promoted by Spire1C and Arp2/3 nucleation factors, and increased DRP1- and MIEF1/2-dependent mitochondrial fission. Changes in mitochondrial dynamics lead to increased production of mitochondrial reactive oxygen species and activate the NRF2 antioxidant transcriptional response, including increased cystine uptake and glutathione metabolism. This retrograde response endows cells with resistance to oxidative stress and reactive oxygen species-dependent chemotherapy drugs. This is relevant in a mouse model of metastatic breast cancer cells dormant in the lung soft tissue, where inhibition of DRP1 and NRF2 restored cisplatin sensitivity and prevented disseminated cancer-cell awakening. We propose that targeting this mitochondrial dynamics- and redox-based mechanotransduction pathway could open avenues to prevent metastatic relapse., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
16. In vitro models of fetal lung development to enhance research into congenital lung diseases.
- Author
-
Shibuya S, Allen-Hyttinen J, De Coppi P, and Michielin F
- Subjects
- Abnormalities, Multiple, Animals, Fetal Development, Fetus, Humans, Lung abnormalities, Mice, Organ Culture Techniques, Organogenesis, Respiration Disorders, Disease Models, Animal, Lung embryology, Lung Diseases metabolism, Morphogenesis
- Abstract
Purpose: This paper aims to build upon previous work to definitively establish in vitro models of murine pseudoglandular stage lung development. These can be easily translated to human fetal lung samples to allow the investigation of lung development in physiologic and pathologic conditions., Methods: Lungs were harvested from mouse embryos at E12.5 and cultured in three different settings, i.e., whole lung culture, mesenchyme-free epithelium culture, and organoid culture. For the whole lung culture, extracted lungs were embedded in Matrigel and incubated on permeable filters. Separately, distal epithelial tips were isolated by firstly removing mesothelial and mesenchymal cells, and then severing the tips from the airway tubes. These were then cultured either in branch-promoting or self-renewing conditions., Results: Cultured whole lungs underwent branching morphogenesis similarly to native lungs. Real-time qPCR analysis demonstrated expression of key genes essential for lung bud formation. The culture condition for epithelial tips was optimized by testing different concentrations of FGF10 and CHIR99021 and evaluating branching formation. The epithelial rudiments in self-renewing conditions formed spherical 3D structures with homogeneous Sox9 expression., Conclusion: We report efficient protocols for ex vivo culture systems of pseudoglandular stage mouse embryonic lungs. These models can be applied to human samples and could be useful to paediatric surgeons to investigate normal lung development, understand the pathogenesis of congenital lung diseases, and explore novel therapeutic strategies.
- Published
- 2021
- Full Text
- View/download PDF
17. NGN2 mmRNA-Based Transcriptional Programming in Microfluidic Guides hiPSCs Toward Neural Fate With Multiple Identities.
- Author
-
Tolomeo AM, Laterza C, Grespan E, Michielin F, Canals I, Kokaia Z, Muraca M, Gagliano O, and Elvassore N
- Abstract
Recent advancements in cell engineering have succeeded in manipulating cell identity with the targeted overexpression of specific cell fate determining transcription factors in a process named transcriptional programming. Neurogenin2 (NGN2) is sufficient to instruct pluripotent stem cells (PSCs) to acquire a neuronal identity when delivered with an integrating system, which arises some safety concerns for clinical applications. A non-integrating system based on modified messenger RNA (mmRNA) delivery method, represents a valuable alternative to lentiviral-based approaches. The ability of NGN2 mmRNA to instruct PSC fate change has not been thoroughly investigated yet. Here we aimed at understanding whether the use of an NGN2 mmRNA-based approach combined with a miniaturized system, which allows a higher transfection efficiency in a cost-effective system, is able to drive human induced PSCs (hiPSCs) toward the neuronal lineage. We show that NGN2 mRNA alone is able to induce cell fate conversion. Surprisingly, the outcome cell population accounts for multiple phenotypes along the neural development trajectory. We found that this mixed population is mainly constituted by neural stem cells (45% ± 18 PAX6 positive cells) and neurons (38% ± 8 βIIITUBULIN positive cells) only when NGN2 is delivered as mmRNA. On the other hand, when the delivery system is lentiviral-based, both providing a constant expression of NGN2 or only a transient pulse, the outcome differentiated population is formed by a clear majority of neurons (88% ± 1 βIIITUBULIN positive cells). Altogether, our data confirm the ability of NGN2 to induce neuralization in hiPSCs and opens a new point of view in respect to the delivery system method when it comes to transcriptional programming applications., 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., (Copyright © 2021 Tolomeo, Laterza, Grespan, Michielin, Canals, Kokaia, Muraca, Gagliano and Elvassore.)
- Published
- 2021
- Full Text
- View/download PDF
18. The Microfluidic Environment Reveals a Hidden Role of Self-Organizing Extracellular Matrix in Hepatic Commitment and Organoid Formation of hiPSCs.
- Author
-
Michielin F, Giobbe GG, Luni C, Hu Q, Maroni I, Orford MR, Manfredi A, Di Filippo L, David AL, Cacchiarelli D, De Coppi P, Eaton S, and Elvassore N
- Subjects
- Cell Differentiation, Hepatocytes metabolism, Humans, Extracellular Matrix metabolism, Liver physiopathology, Microfluidics methods, Organoids physiopathology, Pluripotent Stem Cells metabolism
- Abstract
The specification of the hepatic identity during human liver development is strictly controlled by extrinsic signals, yet it is still not clear how cells respond to these exogenous signals by activating secretory cascades, which are extremely relevant, especially in 3D self-organizing systems. Here, we investigate how the proteins secreted by human pluripotent stem cells (hPSCs) in response to developmental exogenous signals affect the progression from endoderm to the hepatic lineage, including their competence to generate nascent hepatic organoids. By using microfluidic confined environment and stable isotope labeling with amino acids in cell culture-coupled mass spectrometry (SILAC-MS) quantitative proteomic analysis, we find high abundancy of extracellular matrix (ECM)-associated proteins. Hepatic progenitor cells either derived in microfluidics or exposed to exogenous ECM stimuli show a significantly higher potential of forming hepatic organoids that can be rapidly expanded for several passages and further differentiated into functional hepatocytes. These results prove an additional control over the efficiency of hepatic organoid formation and differentiation for downstream applications., Competing Interests: Declaration of Interests D.C. is founder, shareholder, and consultant of Next Generation Diagnostic Srl., (Copyright © 2020. Published by Elsevier Inc.)
- Published
- 2020
- Full Text
- View/download PDF
19. Long-term clinical activity, safety and patient-reported quality of life for emactuzumab-treated patients with diffuse-type tenosynovial giant-cell tumour.
- Author
-
Cassier PA, Italiano A, Gomez-Roca C, Le Tourneau C, Toulmonde M, D'Angelo SP, Weber K, Loirat D, Jacob W, Jegg AM, Michielin F, Christen R, Watson C, Cannarile M, Klaman I, Abiraj K, Ries CH, Weisser M, Rüttinger D, Blay JY, and Delord JP
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Young Adult, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents, Immunological therapeutic use, Quality of Life, Synovitis, Pigmented Villonodular drug therapy
- Abstract
Objectives: This study investigated the safety, clinical activity and patient-reported outcomes of patients with diffuse-type tenosynovial giant-cell tumour (dTGCT) of the soft tissue who were treated with emactuzumab, a humanised anti-colony stimulating factor 1 receptor (CSF1R) monoclonal antibody and were followed up for up to 2 years after the start of treatment., Methods: In this open-label phase 1 study (ClinicalTrials.govNCT01494688), patients received intravenous (IV) emactuzumab from 900 to 2000 mg every two weeks in the dose-escalation phase and at the optimal biological dose of 1000 mg with different schedules in the dose-expansion phase. Adverse event (AE) rates and biomarker assessments from tumour biopsies were analysed. Quality of life was assessed using a standard questionnaire (EuroQol-5D-3L) and the WOMAC® 3.1 Osteoarthritis Index. Tumour responses were determined with magnetic resonance imaging., Results: Altogether, 63 patients were enrolled into the study. The most frequently reported AEs were pruritus, asthenia and oedema. In 36 patients for whom biopsy tissue was available a substantial decrease of CSF1R-positive and CD68/CD163-positive macrophages was detected. The independently reviewed best overall objective response rate (ORR) (Response Evaluation Criteria in Solid Tumors version 1.1) was 71%. Responses were durable, and an ORR of 70% and 64% was determined after one or two years after enrolment into the study. Clinical activity was accompanied by an improvement in EuroQol-5D-3L and particularly the joint disorder-specific WOMAC score., Conclusions: Systemic therapy of dTGCT patients with emactuzumab resulted in pronounced and durable responses associated with symptomatic improvement and a manageable safety profile., (Copyright © 2020 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
20. Phase Ib study of anti-CSF-1R antibody emactuzumab in combination with CD40 agonist selicrelumab in advanced solid tumor patients.
- Author
-
Machiels JP, Gomez-Roca C, Michot JM, Zamarin D, Mitchell T, Catala G, Eberst L, Jacob W, Jegg AM, Cannarile MA, Watson C, Babitzki G, Korski K, Klaman I, Teixeira P, Hoves S, Ries C, Meneses-Lorente G, Michielin F, Christen R, Rüttinger D, Weisser M, Delord JP, and Cassier P
- Subjects
- Antibodies, Monoclonal, Humanized pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Female, Humans, Male, Neoplasms immunology, Receptor, Macrophage Colony-Stimulating Factor metabolism, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, CD40 Antigens metabolism, Neoplasms drug therapy, Receptor, Macrophage Colony-Stimulating Factor antagonists & inhibitors
- Abstract
Background: This phase Ib study evaluated the safety, clinical activity, pharmacokinetics, and pharmacodynamics (PD) of emactuzumab (anti-colony stimulating factor 1 receptor monoclonal antibody (mAb)) in combination with selicrelumab (agonistic cluster of differentiation 40 mAb) in patients with advanced solid tumors., Methods: Both emactuzumab and selicrelumab were administered intravenously every 3 weeks and doses were concomitantly escalated (emactuzumab: 500 to 1000 mg flat; selicrelumab: 2 to 16 mg flat). Dose escalation was conducted using the product of independent beta probabilities dose-escalation design. PD analyzes were performed on peripheral blood samples and tumor/skin biopsies at baseline and on treatment. Clinical activity was evaluated using investigator-based and Response Evaluation Criteria In Solid Tumors V.1.1-based tumor assessments., Results: Three dose-limiting toxicities (all infusion-related reactions (IRRs)) were observed at 8, 12 and 16 mg of selicrelumab together with 1000 mg of emactuzumab. The maximum tolerated dose was not reached at the predefined top doses of emactuzumab (1000 mg) and selicrelumab (16 mg). The most common adverse events were IRRs (75.7%), fatigue (54.1%), facial edema (37.8%), and increase in aspartate aminotransferase and creatinine phosphokinase (35.1% both). PD analyzes demonstrated an increase of Ki67
+ -activated CD8+ T cells accompanied by a decrease of B cells and the reduction of CD14Dim CD16bright monocytes in peripheral blood. The best objective clinical response was stable disease in 40.5% of patients., Conclusion: Emactuzumab in combination with selicrelumab demonstrated a manageable safety profile and evidence of PD activity but did not translate into objective clinical responses., Trialregistration Number: NCT02760797., Competing Interests: Competing interests: Jean-Pascal Machiels: Advisory board consulting for Pfizer, Roche, AstraZeneca, Bayer, Innate, Merck Serono, Boerhinger, BMS, Novartis, Janssen, Incyte, Cue Biopharma and ALX Oncology; travel expenses from Amgen, BMS, Pfizer, MSD; data safety monitoring board support for Debio, Nanobiotix and PsiOxus. Carlos Gomez-Roca: Consultancy for AstraZeneca and BMS; travel grant from Boehringer Ingelheim, BMS, Pierre Fabre, Roche and Sanofi Aventis; honoraria from BMS, Pierre Fabre and Roche; Jean-Marie Michot: Consultancy from Celgene, Bristol Myers Squibb, AstraZeneca and Janssen; travel grant and non-financial support from AstraZeneca, Roche, Novartis, Gilead, Celgene and Bristol Myers Squibb; Dmitriy Zamarin: Consultancy fees from Merck, Synlogic Therapeutics, Biomed Valley Discoveries, Trieza Therapeutics, Tesaro, and Agenus; Tara Mitchell: Advisory board consulting for Merck, BMS and Array; Gaetan Catala: Travel grants from Roche, Pharmamar, MSD and AstraZeneca; advisory role for MSD. Lauriane Eberst: None. Wolfgang Jacob: Sponsor employee and sponsor stock ownership. Anna-Maria Jegg: Former sponsor employee and has patent issued in the use of emactuzumab. Michael A Cannarile: Sponsor employee and sponsor stock ownership. Carl Watson: Sponsor consultant. Galina Babitzki: Sponsor employee. Konstanty Korski: Sponsor employee. Irina Klaman: Sponsor employee. Priscila C Teixeira: Sponsor employee. Sabine Hoves: Sponsor employee, sponsor stock ownership and has patent issued in the use of emactuzumab. Carola Ries: Former sponsor employee and has patent issued in the use of emactuzumab. Georgina Meneses-Lorente: Sponsor employee. Francesca Michielin: Sponsor employee. Randolph Christen: Sponsor employee and sponsor stock ownership. Dominik Rüttinger: Sponsor employee, sponsor stock ownership and has patent issued in the use of emactuzumab. Martin Weisser: Sponsor employee and sponsor stock ownership. Jean-Pierre Delord: Consulting or advisory role for Novartis, Roche/Genentech, Bristol Myers Squibb, MSD Oncology; research funding from Genentech, Bristol Myers Squibb, MSD Oncology. Philippe Cassier: Honoraria from Novartis, Roche/Genentech, Blueprint Medicines, Amgen and AstraZeneca; research funding from Novartis, Roche/Genentech, Eli Lilly, Blueprint Medicines, Bayer, AstraZeneca, Celgene, Plexxikon, AbbVie, Bristol Myers Squibb, Merck Serono, Merck Sharp & Dohme, Taiho Pharmaceuticals, Toray Industries, Transgene, Loxo, GlaxoSmithKline, Innatre Pharma and Janssen; travel grants from Roche, Amgen, Novartis, Bristol Myers Squibb, Merck Sharp & Dohme and Netris Pharma., (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2020
- Full Text
- View/download PDF
21. Therapeutically-induced stable disease in oncology early clinical trials.
- Author
-
Mercier F, Meneses-Lorente G, Grimsey P, Phipps A, and Michielin F
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Models, Biological, Neoplasms pathology, Tumor Burden, Young Adult, Clinical Trials as Topic, Neoplasms drug therapy
- Abstract
Rationale: The RECIST guideline defines four categories of response to treatment for cancer patients according to post-baseline changes in tumor burden, hence ignoring disease history. However, if left untreated, tumors grow exponentially, implying that pretreatment changes in tumor size are key to thoroughly assess efficacy. We present a model-based approach to estimate the rates of changes in tumor mass, before and after treatment onset., Methods: Sixty-eight patients were eligible for the analysis of tumor size data from a Phase 1 study evaluating the effect of emactuzumab. In addition to tumor size measured at baseline and every six weeks during treatment, a pre-baseline measurement was gathered for each patient. A longitudinal regression model was used to estimate the rates of tumor size change before and after treatment onset., Results: The median pre-treatment tumor growth exponential rate was equal to 0.022 month-1, corresponding to a tumor size doubling time of 4 months, and the on-treatment median tumor shrinkage exponential rate was equal to 0.001 month-1. Among sixteen patients categorized as stable disease per RECIST, only five had similar slopes before and after treatment while nine actually improved. One patient in particular had a therapeutically induced stabilization of the disease., Conclusion: Our analysis emphasizes the importance of collecting pre-baseline scans to distinguish therapeutically induced stable disease from cases where the tumor growth is not perturbed by treatment., Competing Interests: The authors have read the journal’s policy and have the following conflicts: all authors are Roche Products Ltd employees and stock holders. This does not alter our adherence to all the PLOS ONE policies on sharing data and materials.
- Published
- 2020
- Full Text
- View/download PDF
22. Extracellular matrix hydrogel derived from decellularized tissues enables endodermal organoid culture.
- Author
-
Giobbe GG, Crowley C, Luni C, Campinoti S, Khedr M, Kretzschmar K, De Santis MM, Zambaiti E, Michielin F, Meran L, Hu Q, van Son G, Urbani L, Manfredi A, Giomo M, Eaton S, Cacchiarelli D, Li VSW, Clevers H, Bonfanti P, Elvassore N, and De Coppi P
- Subjects
- Animals, Cell Proliferation, Endoderm metabolism, Extracellular Matrix chemistry, Humans, Hydrogels chemistry, Hydrogels metabolism, Organoids metabolism, Swine, Tissue Engineering instrumentation, Tissue Scaffolds chemistry, Endoderm growth & development, Extracellular Matrix metabolism, Organoids growth & development
- Abstract
Organoids have extensive therapeutic potential and are increasingly opening up new avenues within regenerative medicine. However, their clinical application is greatly limited by the lack of effective GMP-compliant systems for organoid expansion in culture. Here, we envisage that the use of extracellular matrix (ECM) hydrogels derived from decellularized tissues (DT) can provide an environment capable of directing cell growth. These gels possess the biochemical signature of tissue-specific ECM and have the potential for clinical translation. Gels from decellularized porcine small intestine (SI) mucosa/submucosa enable formation and growth of endoderm-derived human organoids, such as gastric, hepatic, pancreatic, and SI. ECM gels can be used as a tool for direct human organoid derivation, for cell growth with a stable transcriptomic signature, and for in vivo organoid delivery. The development of these ECM-derived hydrogels opens up the potential for human organoids to be used clinically.
- Published
- 2019
- Full Text
- View/download PDF
23. Phase I study of emactuzumab single agent or in combination with paclitaxel in patients with advanced/metastatic solid tumors reveals depletion of immunosuppressive M2-like macrophages.
- Author
-
Gomez-Roca CA, Italiano A, Le Tourneau C, Cassier PA, Toulmonde M, D'Angelo SP, Campone M, Weber KL, Loirat D, Cannarile MA, Jegg AM, Ries C, Christen R, Meneses-Lorente G, Jacob W, Klaman I, Ooi CH, Watson C, Wonde K, Reis B, Michielin F, Rüttinger D, Delord JP, and Blay JY
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Dose-Response Relationship, Drug, Female, Humans, Macrophage Colony-Stimulating Factor blood, Macrophage Colony-Stimulating Factor metabolism, Macrophages immunology, Male, Maximum Tolerated Dose, Middle Aged, Neoplasms blood, Neoplasms immunology, Neoplasms pathology, Paclitaxel therapeutic use, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor antagonists & inhibitors, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Skin cytology, Skin immunology, Treatment Outcome, Young Adult, Antibodies, Monoclonal, Humanized pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Macrophages drug effects, Neoplasms drug therapy, Paclitaxel pharmacology
- Abstract
Background: Emactuzumab is a monoclonal antibody against the colony-stimulating factor-1 receptor and targets tumor-associated macrophages (TAMs). This study assessed the safety, clinical activity, pharmacokinetics (PK) and pharmacodynamics (PD) of emactuzumab, as monotherapy and in combination with paclitaxel, in patients with advanced solid tumors., Patients and Methods: This open-label, phase Ia/b study comprised two parts (dose escalation and dose expansion), each containing two arms (emactuzumab, every 2 or 3 weeks, as monotherapy or in combination with paclitaxel 80 mg/m2 weekly). The dose-escalation part explored the maximum tolerated dose and optimal biological dose (OBD). The dose-expansion part extended the safety assessment and investigated the objective response rate. A PK/PD analysis of serial blood, skin and tumor biopsies was used to explore proof of mechanism and confirm the OBD., Results: No maximum tolerated dose was reached in either study arm, and the safety profile of emactuzumab alone and in combination does not appear to preclude its use. No patients receiving emactuzumab monotherapy showed an objective response; the objective response rate for emactuzumab in combination with paclitaxel was 7% across all doses. Skin macrophages rather than peripheral blood monocytes or circulating colony-stimulating factor-1 were identified as an optimal surrogate PD marker to select the OBD. Emactuzumab treatment alone and in combination with paclitaxel resulted in a plateau of immunosuppressive TAM reduction at the OBD of 1000 mg administered every 2 weeks., Conclusions: Emactuzumab showed specific reduction of immunosuppressive TAMs at the OBD in both treatment arms but did not result in clinically relevant antitumor activity alone or in combination with paclitaxel. (ClinicalTrials.gov Identifier: NCT01494688)., (© The Author(s) 2019. Published by Oxford University Press on behalf of the European Society for Medical Oncology. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
24. A phase Ib/II study of HER3-targeting lumretuzumab in combination with carboplatin and paclitaxel as first-line treatment in patients with advanced or metastatic squamous non-small cell lung cancer.
- Author
-
Cejalvo JM, Jacob W, Fleitas Kanonnikoff T, Felip E, Navarro Mendivil A, Martinez Garcia M, Taus Garcia A, Leighl N, Lassen U, Mau-Soerensen M, Adessi C, Michielin F, James I, Ceppi M, Hasmann M, Weisser M, and Cervantes A
- Abstract
Purpose: This study investigated the safety and clinical activity of lumretuzumab, a humanised antihuman epidermal growth factor receptor 3 (HER3) monoclonal antibody, in combination with carboplatin and paclitaxel in first-line treatment of patients with squamous non-small cell lung cancer (sqNSCLC). HER3 ligand heregulin and HER3 protein expression were evaluated as potential biomarkers of clinical activity., Patients and Methods: This open-label, phase Ib/II study enrolled patients receiving lumretuzumab at 800 mg (flat) in combination with carboplatin (area under the curve (AUC) 6 mg/mL×min) and paclitaxel (200 mg/m
2 ) administered intravenously on a every 3-week schedule. Adverse event (AE) rates and tumour responses were determined. Heregulin messenger RNA (mRNA) and HER3 protein expression were investigated in archival tumour biopsies., Results: Altogether, 12 patients received lumretuzumab in combination with carboplatin and paclitaxel. The most frequent AEs were gastrointestinal, haematological and nervous system toxicities, which were generally mild and manageable. Partial responses were observed in 3 of 12 patients lasting 81, 177 and 207 days. All responses were achieved in tumours expressing higher heregulin mRNA levels., Conclusion: Lumretuzumab in combination with carboplatin and paclitaxel was well tolerated. Objective responses were enriched in tumours expressing higher heregulin mRNA levels., Competing Interests: Competing interests: WJ, MC, MH and MW are the sponsor employees and have sponsor stock ownership. CA and FM are also the sponsor employees. IJ is the sponsor consultant from A4P. AC is the member of the Speaker Bureau of Roche and got research support from Roche.- Published
- 2019
- Full Text
- View/download PDF
25. A Novel Microfluidic Platform for Biomechano-Stimulations on a Chip.
- Author
-
Prevedello L, Michielin F, Balcon M, Savio E, Pavan P, and Elvassore N
- Subjects
- Biomechanical Phenomena, Cells, Cultured, Epithelial Cells physiology, Fibroblasts physiology, Finite Element Analysis, Hot Temperature, Humans, Physical Stimulation, Stress, Mechanical, Lab-On-A-Chip Devices
- Abstract
Mechanical stress has been proven to be an important factor interfering with many biological functions through mechano-sensitive elements within the cells. Despite the current interest in mechano-transduction, the development of suitable experimental tools is still characterized by the strife to design a compact device that allows high-magnification real-time imaging of the stretched cells, thus enabling to follow the dynamics of cellular response to mechanical stimulations. Here we present a microfluidic multi-layered chip that allows mechanical deformation of adherent cells maintaining a fixed focal plane, while allowing independent control of the soluble microenvironment. The device was optimized with the aid of FEM simulation and fully characterized in terms of mechanical deformation. Different cell lines were exposed to tunable mechanical strain, which results in continuous area deformation up to 20%. Thanks to the coupling of chemical glass etching, 2-dimensional deformation of a thin elastomeric membrane and microfluidic cell culture, the developed device allows a unique combination of cell mechanical stimulation, in line imaging and accurate control of cell culture microenvironment.
- Published
- 2019
- Full Text
- View/download PDF
26. Phase Ib study evaluating safety and clinical activity of the anti-HER3 antibody lumretuzumab combined with the anti-HER2 antibody pertuzumab and paclitaxel in HER3-positive, HER2-low metastatic breast cancer.
- Author
-
Schneeweiss A, Park-Simon TW, Albanell J, Lassen U, Cortés J, Dieras V, May M, Schindler C, Marmé F, Cejalvo JM, Martinez-Garcia M, Gonzalez I, Lopez-Martin J, Welt A, Levy C, Joly F, Michielin F, Jacob W, Adessi C, Moisan A, Meneses-Lorente G, Racek T, James I, Ceppi M, Hasmann M, Weisser M, and Cervantes A
- Subjects
- Adult, Aged, Antibodies, Monoclonal, Humanized adverse effects, Antibodies, Monoclonal, Humanized pharmacokinetics, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Antineoplastic Combined Chemotherapy Protocols adverse effects, Breast Neoplasms genetics, Breast Neoplasms metabolism, Diarrhea chemically induced, Female, Humans, Hypokalemia chemically induced, Middle Aged, Paclitaxel adverse effects, Polymorphism, Single Nucleotide, Receptor, ErbB-2 genetics, Receptor, ErbB-2 metabolism, Receptor, ErbB-3 antagonists & inhibitors, Receptor, ErbB-3 genetics, Receptor, ErbB-3 metabolism, Antibodies, Monoclonal, Humanized administration & dosage, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Breast Neoplasms drug therapy, Paclitaxel administration & dosage, Receptor, ErbB-2 antagonists & inhibitors
- Abstract
Purpose To investigate the safety and clinical activity of comprehensive human epidermal growth factor receptor (HER) family receptor inhibition using lumretuzumab (anti-HER3) and pertuzumab (anti-HER2) in combination with paclitaxel in patients with metastatic breast cancer (MBC). Methods This phase Ib study enrolled 35 MBC patients (first line or higher) with HER3-positive and HER2-low (immunohistochemistry 1+ to 2+ and in-situ hybridization negative) tumors. Patients received lumretuzumab (1000 mg in Cohort 1; 500 mg in Cohorts 2 and 3) plus pertuzumab (840 mg loading dose [LD] followed by 420 mg in Cohorts 1 and 2; 420 mg without LD in Cohort 3) every 3 weeks, plus paclitaxel (80 mg/m
2 weekly in all cohorts). Patients in Cohort 3 received prophylactic loperamide treatment. Results Diarrhea grade 3 was a dose-limiting toxicity of Cohort 1 defining the maximum tolerated dose of lumretuzumab when given in combination with pertuzumab and paclitaxel at 500 mg every three weeks. Grade 3 diarrhea decreased from 50% (Cohort 2) to 30.8% (Cohort 3) with prophylactic loperamide administration and omission of the pertuzumab LD, nonetheless, all patients still experienced diarrhea. In first-line MBC patients, the objective response rate in Cohorts 2 and 3 was 55% and 38.5%, respectively. No relationship between HER2 and HER3 expression or somatic mutations and clinical response was observed. Conclusions Combination treatment with lumretuzumab, pertuzumab and paclitaxel was associated with a high incidence of diarrhea. Despite the efforts to alter dosing, the therapeutic window remained too narrow to warrant further clinical development., Trial Registration: on ClinicalTrials.gov with the identifier NCT01918254 first registered on 3rd July 2013.- Published
- 2018
- Full Text
- View/download PDF
27. Mechanistic Investigations of Diarrhea Toxicity Induced by Anti-HER2/3 Combination Therapy.
- Author
-
Moisan A, Michielin F, Jacob W, Kronenberg S, Wilson S, Avignon B, Gérard R, Benmansour F, McIntyre C, Meneses-Lorente G, Hasmann M, Schneeweiss A, Weisser M, and Adessi C
- Subjects
- Adult, Aged, Antibodies, Anti-Idiotypic adverse effects, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols adverse effects, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology, Caco-2 Cells, Cell Proliferation drug effects, Colonic Neoplasms genetics, Colonic Neoplasms pathology, Combined Modality Therapy, Diarrhea chemically induced, Diarrhea pathology, Dose-Response Relationship, Drug, Drug-Related Side Effects and Adverse Reactions drug therapy, Drug-Related Side Effects and Adverse Reactions pathology, Female, Humans, Intestinal Mucosa drug effects, Intestinal Mucosa pathology, Middle Aged, Neoplasm Metastasis, Paclitaxel administration & dosage, Paclitaxel adverse effects, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-3 antagonists & inhibitors, Antibodies, Anti-Idiotypic administration & dosage, Colonic Neoplasms drug therapy, Diarrhea genetics, Receptor, ErbB-2 genetics, Receptor, ErbB-3 genetics
- Abstract
Combination of targeted therapies is expected to provide superior efficacy in the treatment of cancer either by enhanced antitumor activity or by preventing or delaying the development of resistance. Common challenges in developing combination therapies include the potential of additive and aggravated toxicities associated with pharmacologically related adverse effects. We have recently reported that combination of anti-HER2 and anti-HER3 antibodies, pertuzumab and lumretuzumab, along with paclitaxel chemotherapy in metastatic breast cancer, resulted in a high incidence of diarrhea that ultimately limited further clinical development of this combination. Here, we further dissected the diarrhea profile of the various patient dose cohorts and carried out in vitro investigations in human colon cell lines and explants to decipher the contribution and the mechanism of anti-HER2/3 therapeutic antibodies to intestinal epithelium malfunction. Our clinical investigations in patients revealed that while dose reduction of lumretuzumab, omission of pertuzumab loading dose, and introduction of a prophylactic antidiarrheal treatment reduced most severe adverse events, patients still suffered from persistent diarrhea during the treatment. Our in vitro investigations showed that pertuzumab and lumretuzumab combination treatment resulted in upregulation of chloride channel activity without indication of intestinal barrier disruption. Overall, our findings provide a mechanistic rationale to explore alternative of conventional antigut motility using medication targeting chloride channel activity to mitigate diarrhea of HER combination therapies. Mol Cancer Ther; 17(7); 1464-74. ©2018 AACR ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
28. Phase Ib Study of Lumretuzumab Plus Cetuximab or Erlotinib in Solid Tumor Patients and Evaluation of HER3 and Heregulin as Potential Biomarkers of Clinical Activity.
- Author
-
Meulendijks D, Jacob W, Voest EE, Mau-Sorensen M, Martinez-Garcia M, Taus A, Fleitas T, Cervantes A, Lolkema MP, Langenberg MHG, De Jonge MJ, Sleijfer S, Han JY, Calles A, Felip E, Kim SW, Schellens JHM, Wilson S, Thomas M, Ceppi M, Meneses-Lorente G, James I, Vega-Harring S, Dua R, Nguyen M, Steiner L, Adessi C, Michielin F, Bossenmaier B, Weisser M, and Lassen UN
- Subjects
- Antibodies, Monoclonal, Humanized administration & dosage, Antineoplastic Combined Chemotherapy Protocols adverse effects, Cetuximab administration & dosage, Erlotinib Hydrochloride administration & dosage, Female, Gene Expression Regulation, Neoplastic, Humans, Male, Neoplasms mortality, Neoplasms pathology, Neuregulin-1 genetics, Receptor, ErbB-3 genetics, Survival Analysis, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor, Neoplasms drug therapy, Neoplasms metabolism, Neuregulin-1 metabolism, Receptor, ErbB-3 metabolism
- Abstract
Purpose: This study investigated the safety, clinical activity, and target-associated biomarkers of lumretuzumab, a humanized, glycoengineered, anti-HER3 monoclonal antibody (mAb), in combination with the EGFR-blocking agents erlotinib or cetuximab in patients with advanced HER3-positive carcinomas. Experimental Design: The study included two parts: dose escalation and dose extension phases with lumretuzumab in combination with either cetuximab or erlotinib, respectively. In both parts, patients received lumretuzumab doses from 400 to 2,000 mg plus cetuximab or erlotinib according to standard posology, respectively. The effect of HRG mRNA and HER3 mRNA and protein expression were investigated in a dedicated extension cohort of squamous non-small cell lung cancer (sqNSCLC) patients treated with lumretuzumab and erlotinib. Results: Altogether, 120 patients were treated. One dose-limiting toxicity (DLT) in the cetuximab part and two DLTs in the erlotinib part were reported. The most frequent adverse events were gastrointestinal and skin toxicities, which were manageable. The objective response rate (ORR) was 6.1% in the cetuximab part and 4.2% in the erlotinib part. In the sqNSCLC extension cohort of the erlotinib part, higher tumor HRG and HER3 mRNA levels were associated with a numerically higher disease control rate but not ORR. Conclusions: The toxicity profile of lumretuzumab in combination with cetuximab and erlotinib was manageable, but only modest clinical activity was observed across tumor types. In the sqNSCLC cohort, there was no evidence of meaningful clinical benefit despite enriching for tumors with higher HRG mRNA expression levels. Clin Cancer Res; 23(18); 5406-15. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
29. Phase I clinical study of RG7356, an anti-CD44 humanized antibody, in patients with acute myeloid leukemia.
- Author
-
Vey N, Delaunay J, Martinelli G, Fiedler W, Raffoux E, Prebet T, Gomez-Roca C, Papayannidis C, Kebenko M, Paschka P, Christen R, Guarin E, Bröske AM, Baehner M, Brewster M, Walz AC, Michielin F, Runza V, Meresse V, and Recher C
- Subjects
- Adult, Aged, Aged, 80 and over, Dose-Response Relationship, Drug, Female, Humans, Leukemia, Myeloid, Acute pathology, Male, Middle Aged, Young Adult, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Leukemia, Myeloid, Acute drug therapy
- Abstract
RG7356, a recombinant anti-CD44 immunoglobulin G1 humanized monoclonal antibody, inhibits cell adhesion and has been associated with macrophage activation in preclinical models. We report results of a phase I dose-escalation study of RG7356 in relapsed/refractory acute myeloid leukemia (AML).Eligible patients with refractory AML, relapsed AML after induction chemotherapy, or previously untreated AML not eligible for intensive chemotherapy were enrolled and received intravenous RG7356 at dosages ≤ 2400 mg every other week or ≤ 1200 mg weekly or twice weekly; dose escalation started at 300 mg.Forty-four patients (median age, 69 years) were enrolled. One dose-limiting toxicity occurred (grade 3 hemolysis exacerbation) after one 1200 mg dose (twice-weekly cohort). The majority of adverse events were mild/moderate. Infusion-related reactions occurred in 64% of patients mainly during cycle 1. Two patients experienced grade 3 drug-induced aseptic meningitis. Pharmacokinetics increased supraproportionally, suggesting a target-mediated drug disposition (TMDD) at ≥ 1200 mg. Two patients achieved complete response with incomplete platelet recovery or partial response, respectively. One patient had stable disease with hematologic improvement.RG7356 was generally safe and well tolerated. Maximum tolerated dose was not reached, but saturation of TMDD was achieved. The recommended dose for future AML evaluations is 2400 mg every other week., Competing Interests: N. Vey: Roche (honoraria); J. Delaunay: No conflicts to disclose; G. Martinelli: Speakers’ bureau: Novartis, BMS, Celgene; Consultant: Ariad, Pfizer, Roche, MSD; W. Fiedler: Research funding: Pfizer Pharma GmbH; Travel grants: Teva GmbH, Gilead Sciences GmbH; E. Raffoux: No conflicts to disclose; T. Prebet: No conflicts to disclose; C. Gomez-Roca: No conflicts to disclose; C. Papayanndis: No conflicts to disclose; M. Kebenko: No conflicts to disclose; P. Paschka: No conflicts to disclose; C. Recher: Research funding: Amgen, Chugai, Novartis, Celgene; Advisory board: Celgene, Sunesis; R. Christen, E. Guarin, A-B. Bröske, M. Baehner, M. Brewster, A-C. Walz, F. Michielin, V. Runza, and V. Meresse are or have been employees of Roche.
- Published
- 2016
- Full Text
- View/download PDF
30. High-efficiency cellular reprogramming with microfluidics.
- Author
-
Luni C, Giulitti S, Serena E, Ferrari L, Zambon A, Gagliano O, Giobbe GG, Michielin F, Knöbel S, Bosio A, and Elvassore N
- Subjects
- Cells, Cultured, Fibroblasts cytology, Humans, RNA, Messenger genetics, Cellular Reprogramming genetics, Cellular Reprogramming Techniques methods, Induced Pluripotent Stem Cells cytology, Microfluidics methods
- Abstract
We report that the efficiency of reprogramming human somatic cells to induced pluripotent stem cells (hiPSCs) can be dramatically improved in a microfluidic environment. Microliter-volume confinement resulted in a 50-fold increase in efficiency over traditional reprogramming by delivery of synthetic mRNAs encoding transcription factors. In these small volumes, extracellular components of the TGF-β and other signaling pathways exhibited temporal regulation that appears critical to acquisition of pluripotency. The high quality and purity of the resulting hiPSCs (μ-hiPSCs) allowed direct differentiation into functional hepatocyte- and cardiomyocyte-like cells in the same platform without additional expansion.
- Published
- 2016
- Full Text
- View/download PDF
31. First-in-Human Phase I Study of Lumretuzumab, a Glycoengineered Humanized Anti-HER3 Monoclonal Antibody, in Patients with Metastatic or Advanced HER3-Positive Solid Tumors.
- Author
-
Meulendijks D, Jacob W, Martinez-Garcia M, Taus A, Lolkema MP, Voest EE, Langenberg MH, Fleitas Kanonnikoff T, Cervantes A, De Jonge MJ, Sleijfer S, Soerensen MM, Thomas M, Ceppi M, Meneses-Lorente G, James I, Adessi C, Michielin F, Abiraj K, Bossenmaier B, Schellens JH, Weisser M, and Lassen UN
- Subjects
- Adult, Aged, Analgesics pharmacology, Antibodies, Monoclonal, Humanized pharmacology, Colorectal Neoplasms metabolism, Female, Humans, Male, Maximum Tolerated Dose, Middle Aged, Receptor, ErbB-3 antagonists & inhibitors, Receptor, ErbB-3 metabolism, Treatment Outcome, Analgesics therapeutic use, Antibodies, Monoclonal, Humanized therapeutic use, Colorectal Neoplasms drug therapy
- Abstract
Purpose: A first-in-human phase I study was conducted to characterize safety, efficacy, and pharmacokinetic (PK) and pharmacodynamic (PD) properties of lumretuzumab, a humanized and glycoengineered anti-HER3 monoclonal antibody, in patients with advanced cancer., Experimental Design: Twenty-five patients with histologically confirmed HER3-expressing tumors received lumretuzumab (100, 200, 400, 800, 1,600, and 2,000 mg) every two weeks (q2w) in 3+3 dose-escalation phase. In addition, 22 patients were enrolled into an extension cohort at 2,000 mg q2w., Results: There were no dose-limiting toxicities. Common adverse events (any grade) included diarrhea (22 patients, 46.8%), fatigue (21 patients, 44.7%), decreased appetite (15 patients, 31.9%), infusion-related reactions (13 patients, 27.7%), and constipation (10 patients, 21.3%). The peak concentration (Cmax) and area under the concentration-time curve up to the last measurable concentration (AUClast) of lumretuzumab increased more than dose proportionally from 100 mg up to 400 mg. Linear PK was observed with doses ≥ 400 mg q2w indicating target-mediated drug disposition saturation. Downregulation of HER3 membranous protein was observed in on-treatment tumor biopsies from 200 mg, and was maximal at and above 400 mg. An ex vivo assay demonstrated increased activation potential of peripheral NK lymphocytes with lumretuzumab compared with a non-glycoengineered anti-HER3 antibody. Ten patients (21.3%) had stable disease and remained on study at a median of 111 days (range, 80-225 days)., Conclusions: Lumretuzumab was well tolerated and showed evidence of clinical activity. Linear serum PK properties and plateauing of PD effects in serial tumor biopsies indicate optimal biologically active doses of lumretuzumab from 400 mg onwards., (©2015 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
32. NUTRITIONAL AND METABOLIC ASSESSMENT IN OVERWEIGHT PATIENTS WITH AND WITHOUT HYPERPROLACTINEMIA CAUSED BY PROLACTINOMA.
- Author
-
Breyer Freitas B, Rothen RE, Zeni D, Garcia Soares Leães C, Costa Oliveira M, Michielin Busnello F, and Fernanda Semmelmann Pereira-Lima J
- Subjects
- Adult, Aged, Cross-Sectional Studies, Diet, Eating, Feeding Behavior, Female, Humans, Male, Middle Aged, Nutritional Status, Obesity etiology, Obesity metabolism, Hyperprolactinemia etiology, Hyperprolactinemia metabolism, Nutrition Assessment, Overweight complications, Overweight metabolism, Pituitary Neoplasms complications, Pituitary Neoplasms metabolism, Prolactinoma complications, Prolactinoma metabolism
- Abstract
Introduction: prolactinomas are pituitary adenomas that express and secrete prolactin. These patients are overweight and the mechanisms are being studied., Goals: assess nutritional and metabolic status of overweight patients with and without hyperprolactinemia caused by prolactinoma and compare them., Materials and Methods: cross-sectional study, patients with body mass index (BMI) ≥ 25 kg/m2 with and without prolactinoma: 1) 20 normoprolactinemic (NPrl) with prolactinoma; 2) 23 hyperprolactinemic (HPrl) with prolactinoma; 3) 28 controls without prolactinoma or alterations in prolactin levels. Evaluated through anthropometric, dietetics, and biochemical assessment., Results: of the 71 patients evaluated, most were obese women with macroprolactinomas. All three groups had diets with low caloric and monounsaturated fatty acid (MUFA) intake, the NPrl group had low carbohydrate (CHO) intake and high lipid (LIP) and saturated fatty acid (SFA) intake, and the NPrl and HPrl groups had appropriate intake of polyunsaturated fatty acids (PUFA). The HPrl group had elevated total cholesterol. HDL cholesterol was below the recommended threshold for most patients. No statistically significant differences were found in anthropometric and biochemical variables among the groups., Conclusions: most patients with prolactinomas and controls are obese and metabolically similar regardless of prolactin levels. All groups presented low caloric and MUFA intake. Protein, LIP, SFA, and cholesterol were significantly different among the groups, the NPrl group ingested less amount of protein and greater of fat. Snacking between meals and changes of food consumption on weekends was reported by most patients. This is the first study comparing patients with prolactinomas and controls, both with overweight, regarding food consumption and feeding behavior., (Copyright AULA MEDICA EDICIONES 2014. Published by AULA MEDICA. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
33. Functional differentiation of human pluripotent stem cells on a chip.
- Author
-
Giobbe GG, Michielin F, Luni C, Giulitti S, Martewicz S, Dupont S, Floreani A, and Elvassore N
- Subjects
- Cell Differentiation, Embryonic Stem Cells cytology, Hepatocytes cytology, Humans, Myocytes, Cardiac cytology, Microfluidic Analytical Techniques methods, Pluripotent Stem Cells cytology
- Abstract
Microengineering human "organs-on-chips" remains an open challenge. Here, we describe a robust microfluidics-based approach for the differentiation of human pluripotent stem cells directly on a chip. Extrinsic signal modulation, achieved through optimal frequency of medium delivery, can be used as a parameter for improved germ layer specification and cell differentiation. Human cardiomyocytes and hepatocytes derived on chips showed functional phenotypes and responses to temporally defined drug treatments.
- Published
- 2015
- Full Text
- View/download PDF
34. A mechanical checkpoint controls multicellular growth through YAP/TAZ regulation by actin-processing factors.
- Author
-
Aragona M, Panciera T, Manfrin A, Giulitti S, Michielin F, Elvassore N, Dupont S, and Piccolo S
- Subjects
- Actins metabolism, Acyltransferases, Adaptor Proteins, Signal Transducing antagonists & inhibitors, Breast Neoplasms pathology, Cell Line, Tumor, Extracellular Matrix metabolism, Humans, Mechanical Phenomena, Phosphoproteins antagonists & inhibitors, Transcription Factors antagonists & inhibitors, YAP-Signaling Proteins, Actin Capping Proteins metabolism, Adaptor Proteins, Signal Transducing metabolism, Breast Neoplasms metabolism, Cell Proliferation, Phosphoproteins metabolism, Signal Transduction, Transcription Factors metabolism
- Abstract
Key cellular decisions, such as proliferation or growth arrest, typically occur at spatially defined locations within tissues. Loss of this spatial control is a hallmark of many diseases, including cancer. Yet, how these patterns are established is incompletely understood. Here, we report that physical and architectural features of a multicellular sheet inform cells about their proliferative capacity through mechanical regulation of YAP and TAZ, known mediators of Hippo signaling and organ growth. YAP/TAZ activity is confined to cells exposed to mechanical stresses, such as stretching, location at edges/curvatures contouring an epithelial sheet, or stiffness of the surrounding extracellular matrix. We identify the F-actin-capping/severing proteins Cofilin, CapZ, and Gelsolin as essential gatekeepers that limit YAP/TAZ activity in cells experiencing low mechanical stresses, including contact inhibition of proliferation. We propose that mechanical forces are overarching regulators of YAP/TAZ in multicellular contexts, setting responsiveness to Hippo, WNT, and GPCR signaling., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
35. Stochastic model-assisted development of efficient low-dose viral transduction in microfluidics.
- Author
-
Luni C, Michielin F, Barzon L, Calabrò V, and Elvassore N
- Subjects
- Fibroblasts metabolism, Fibroblasts virology, Foreskin cytology, Gene Expression, Genetic Vectors, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Humans, Male, Microscopy, Fluorescence, Stochastic Processes, Adenoviridae genetics, Microfluidics, Models, Genetic, Transduction, Genetic
- Abstract
Adenoviruses are commonly used in vitro as gene transfer vectors in multiple applications. Nevertheless, issues such as low infection efficiency and toxicity effects on host cells have not been resolved yet. This work aims at developing a new versatile tool to enhance the expression of transduced genes while working at low viral doses in a sequential manner. We developed a microfluidic platform with automatically controlled sequential perfusion stages, which includes 10 independent channels. In addition, we built a stochastic mathematical model, accounting for the discrete nature of cells and viruses, to predict not only the percentage of infected cells, but also the associated infecting-virus distribution in the cell population. Microfluidic system and mathematical model were coupled to define an efficient experimental strategy. We used human foreskin fibroblasts, infected by replication-incompetent adenoviruses carrying EGFP gene, as the testing system. Cell characterization was performed through fluorescence microscopy, followed by image analysis. We explored the effect of different aspects: perfusion, multiplicity of infection, and temporal patterns of infection. We demonstrated feasibility of performing efficient viral transduction at low doses, by repeated pulses of cell-virus contact. This procedure also enhanced the exogenous gene expression in the sequential microfluidic infection system compared to a single infection at a higher, nontoxic, viral dose., (Copyright © 2013 Biophysical Society. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
36. Slow dissociation of partial agonists from the D₂ receptor is linked to reduced prolactin release.
- Author
-
Carboni L, Negri M, Michielin F, Bertani S, Fratte SD, Oliosi B, and Cavanni P
- Subjects
- Animals, Aripiprazole, Clozapine pharmacology, Dibenzothiazepines pharmacology, Dopamine D2 Receptor Antagonists, Haloperidol pharmacology, Male, Piperazines pharmacology, Prolactin antagonists & inhibitors, Prolactin blood, Quetiapine Fumarate, Quinolones pharmacology, Rats, Rats, Sprague-Dawley, Receptors, Dopamine D2 agonists, Antipsychotic Agents pharmacology, Dopamine Agonists pharmacology, Dopamine Antagonists pharmacology, Prolactin metabolism, Receptors, Dopamine D2 metabolism
- Abstract
In this study we investigated the correlation between affinity, efficacy, peripheral receptor occupancy, and kinetic properties of D₂ dopamine receptor ligands with time-course evaluations of prolactin release in rat blood. We profiled typical and atypical antipsychotic antagonists at D₂ receptors, the partial agonist aripiprazole, and four novel partial agonist compounds with different properties. Clozapine and quetiapine revealed lower prolactin release and fast dissociation kinetics, linking fast dissociation and prolactin-sparing properties. Surprisingly, haloperidol, a highly prolactin-releasing antagonist, shared intermediate dissociation properties. Factors other than kinetic properties may thus contribute to prolactin-releasing properties of antagonists. Partial agonists sharing similar efficacies and receptor occupancies differed markedly in their ability to induce hyperprolactinaemia. Aripiprazole moderately released prolactin even at high receptor occupancies, with slow dissociation from D₂ receptors. Other compounds displaying low affinities and fast dissociations released prolactin substantially, although less than haloperidol. The effect augmented after repeated administrations. Compounds with high affinities and slow dissociation rates stimulated moderate prolactin release at high receptor occupancies, reaching a ceiling effect at 50-60% occupancy. Moreover, the effect developed tolerance. In conclusion, we investigated the affinity and kinetic properties of D₂ partial agonists associated with their ability to induce prolactin release in blood. We propose that for D₂ partial agonists, at comparable intrinsic activities and peripheral occupancies, the prolactin-releasing properties are linked to their kinetic rate properties. Differently from D₂ antagonists, partial agonists display slow dissociation and high affinity associated with a low prolactin release profile.
- Published
- 2012
- Full Text
- View/download PDF
37. Reversible alteration of calcium dynamics in cardiomyocytes during acute hypoxia transient in a microfluidic platform.
- Author
-
Martewicz S, Michielin F, Serena E, Zambon A, Mongillo M, and Elvassore N
- Subjects
- Animals, Animals, Newborn, Calcium analysis, Cell Hypoxia physiology, Microfluidic Analytical Techniques instrumentation, Microfluidic Analytical Techniques methods, Microscopy, Confocal, Numerical Analysis, Computer-Assisted, Rats, Rats, Sprague-Dawley, Calcium metabolism, Calcium Channels, L-Type metabolism, Myocardium metabolism, Myocytes, Cardiac metabolism
- Abstract
Heart disease is the leading cause of mortality in western countries. Apart from congenital and anatomical alterations, ischemia is the most common agent causing myocardial damage. During ischemia, a sudden decrease in oxygen concentration alters cardiomyocyte function and compromises cell survival. The calcium handling machinery, which regulates the main functional features of a cardiomyocyte, is heavily compromised during acute hypoxic events. Alterations in calcium dynamics have been linked to both short- and long-term consequences of ischemia, ranging from arrhythmias to heart failure. In this perspective, we aimed at investigating the calcium dynamics in functional cardiomyocytes during the early phase of a hypoxic event. For this purpose, we developed a microfluidic system specifically designed for controlling fast oxygen concentration dynamics through a gas micro-exchanger allowing in line analysis of intracellular calcium concentration by confocal microscopy. Experimental results show that exposure of Fluo-4 loaded neonatal rat cardiomyocytes to hypoxic conditions induced changes in intracellular Ca(2+) transients. Such behavior was reversible and was detected for hypoxic levels below 5% of oxygen partial pressure. The observed changes in Ca(2+) dynamics were mimicked using specific L-type Ca(2+) channel antagonists, suggesting that alterations in calcium channel function occur at low oxygen levels. Reversible alteration in ion channel function, that takes place in response to changes in cellular oxygen, might represent an adaptive mechanism of cardiopreservation during ischemia., (This journal is © The Royal Society of Chemistry 2012)
- Published
- 2012
- Full Text
- View/download PDF
38. rKv1.2 overexpression in the central medial thalamic area decreases caffeine-induced arousal.
- Author
-
Cazzin C, Piccoli L, Massagrande M, Garbati N, Michielin F, Knaus HG, Ring CJ, Morrison AD, Merlo-Pich E, Rovo Z, Astori S, Lüthi A, Corti C, and Corsi M
- Subjects
- Animals, Behavior, Animal physiology, Cells, Cultured, Dependovirus genetics, Fluorescent Antibody Technique, Genetic Vectors, Green Fluorescent Proteins genetics, Immunohistochemistry, In Situ Hybridization, Male, Patch-Clamp Techniques, Rats, Sleep genetics, Sleep physiology, Telemetry, Arousal drug effects, Arousal genetics, Caffeine pharmacology, Central Nervous System Stimulants pharmacology, Intralaminar Thalamic Nuclei metabolism, Kv1.2 Potassium Channel genetics
- Abstract
The voltage-gated potassium channel Kv1.2 belongs to the shaker-related family and has recently been implicated in the control of sleep profile on the basis of clinical and experimental evidence in rodents. To further investigate whether increasing Kv1.2 activity would promote sleep occurrence in rats, we developed an adeno-associated viral vector that induces overexpression of rat Kv1.2 protein. The viral vector was first evaluated in vitro for its ability to overexpress rat Kv1.2 protein and to produce functional currents in infected U2OS cells. Next, the adeno-associated Kv1.2 vector was injected stereotaxically into the central medial thalamic area of rats and overexpression of Kv1.2 was showed by in situ hybridization, ex vivo electrophysiology and immunohistochemistry. Finally, the functional effect of Kv1.2 overexpression on sleep facilitation was investigated using telemetry system under normal conditions and following administration of the arousing agent caffeine, during the light phase. While no differences in sleep profile were observed between the control and the treated animals under normal conditions, a decrease in the pro-arousal effect of caffeine was seen only in the animals injected with the adeno-associated virus-Kv1.2 vector. Overall, our data further support a role of the Kv1.2 channel in the control of sleep profile, particularly under conditions of sleep disturbance., (© 2011 The Authors. Genes, Brain and Behavior © 2011 Blackwell Publishing Ltd and International Behavioural and Neural Genetics Society.)
- Published
- 2011
- Full Text
- View/download PDF
39. Effect of the p38 MAPK inhibitor SB-239063 on Lipopolysaccharide-induced psychomotor retardation and peripheral biomarker alterations in rats.
- Author
-
Bison S, Razzoli M, Arban R, Michielin F, Bertani S, and Carboni L
- Subjects
- Animals, Biomarkers metabolism, Hormones metabolism, Imidazoles therapeutic use, Inflammation metabolism, Male, Motor Activity drug effects, Protein Kinase Inhibitors therapeutic use, Psychomotor Disorders immunology, Psychomotor Disorders metabolism, Pyrimidines therapeutic use, Rats, Rats, Sprague-Dawley, Stress, Physiological drug effects, Time Factors, Imidazoles pharmacology, Lipopolysaccharides pharmacology, Protein Kinase Inhibitors pharmacology, Psychomotor Disorders chemically induced, Psychomotor Disorders drug therapy, Pyrimidines pharmacology, p38 Mitogen-Activated Protein Kinases antagonists & inhibitors
- Abstract
Lipopolysaccharide (LPS) administration in rats induces a characteristic syndrome termed 'sickness behavior', including profound changes on locomotor activity and circulating stress and inflammatory mediators. The aim of the present investigation was to evaluate whether the behavioral and the peripheral biomarker responses induced by LPS could be modified by acute treatment with the p38 mitogen-activated protein kinase inhibitor SB-239063. Male Sprague-Dawley rats were treated orally either with vehicle or SB-239063 (3, 10 and 30 mg/kg) 1h before an intraperitoneal injection of either saline or LPS 125 μg/kg. Two hours after LPS injection, rats were placed in a novel open field arena for locomotion assessment during both the light and dark periods. Inflammation and stress mediators were evaluated in plasma 2, 3, 5 or 14 h into the dark phase. Pre-treatment with SB-239063 significantly reversed the locomotor deficits induced by LPS injection. Interleukin (IL)-1β, IL-6, IL-10, Granulocyte-Macrophage-Colony Stimulating Factor, Interferon-γ, and C-reactive-protein levels were increased significantly by LPS, but not when LPS was preceded by SB-239063 treatment. LPS significantly decreased growth-hormone and Prolactin, and this effect was attenuated by SB-239063. Tumor Necrosis Factor-α, Adrenocorticotropic Hormone and Corticosterone levels were significantly higher in LPS-treated rats and were not normalized by SB-239063. Thus, we demonstrate that acute treatment with SB-239063 may have ameliorating effects in early changes of LPS-induced sickness behavior and alteration in the peripheral cytokines/hormones. As such, our procedure may offer an opportunity to test the activity of novel anti-inflammatory compounds on specific symptoms of sickness associated with neuroimmune dysfunctions., (Copyright © 2011 Elsevier B.V. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
40. Altered levels of glutamatergic receptors and Na+/K+ ATPase-α1 in the prefrontal cortex of subjects with schizophrenia.
- Author
-
Corti C, Xuereb JH, Crepaldi L, Corsi M, Michielin F, and Ferraguti F
- Subjects
- Adult, Aged, Aged, 80 and over, Analysis of Variance, Female, Humans, Male, Middle Aged, Postmortem Changes, Receptors, Glutamate classification, Statistics, Nonparametric, Prefrontal Cortex metabolism, Receptors, Glutamate metabolism, Schizophrenia pathology, Sodium-Potassium-Exchanging ATPase metabolism
- Abstract
Evidence has accumulated over the past years that dysregulation of glutamatergic neurotransmission maybe implicated in the pathophysiology of schizophrenia. Glutamate acts on two major classes of receptors: ionotropic receptors, which are ligand-gated ion channels, and metabotropic receptors (mGluRs), coupled to heterotrimeric G-proteins. Although several pharmacological evidences point to abnormal glutamatergic transmission in schizophrenia, changes in the expression of glutamatergic receptors in the prefrontal cortex of patients with schizophrenia remains equivocal. In the present work, we have investigated glutamatergic neurotransmission in schizophrenia by assessing the expression in Brodmann Area 10 of mGluR5, the AMPA receptor subunits GluR1 and GluR2, and Na(+)/K(+) ATPase-α1, a potential modulator of glutamate uptake in the brain. Semiquantitative analysis of the expression of these proteins from postmortem brains revealed a particularly prominent reduction of GluR1 and GluR2 expression in patients with schizophrenia vs the control group. Conversely, we observed an up-regulation in the levels of Na(+)/K(+) ATPase-α1 expression. Finally, no change in the protein levels of mGluR5 was observed in schizophrenia. Our findings support and expand the hypothesis of glutamatergic dysfunction in prefrontal cortex in the pathophysiology of schizophrenia., (Copyright © 2011 Elsevier B.V. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
41. Macrolide antibiotics broadly and distinctively inhibit cytokine and chemokine production by COPD sputum cells in vitro.
- Author
-
Marjanović N, Bosnar M, Michielin F, Willé DR, Anić-Milić T, Culić O, Popović-Grle S, Bogdan M, Parnham MJ, and Eraković Haber V
- Subjects
- Azithromycin pharmacology, Cell Survival drug effects, Cells, Cultured, Clarithromycin pharmacology, Erythromycin pharmacology, Humans, Pulmonary Disease, Chronic Obstructive immunology, Roxithromycin pharmacology, Sputum immunology, Anti-Bacterial Agents pharmacology, Chemokines immunology, Cytokines immunology, Macrolides pharmacology, Pulmonary Disease, Chronic Obstructive drug therapy, Sputum cytology
- Abstract
Macrolide antibiotics are known to exert anti-inflammatory actions in vivo, including certain effects in COPD patients. In order to investigate the immunomodulatory profile of activity of macrolide antibiotics, we have studied the effects of azithromycin, clarithromycin, erythromycin and roxithromycin on the in vitro production of a panel of inflammatory mediators from cells isolated from human, steroid-naïve, COPD sputum samples. Macrolide effects were compared to three other commonly used anti-inflammatory compounds, the corticosteroid dexamethasone, the PDE4 inhibitor, roflumilast and the p38 kinase inhibitor, SB203580. Three of the four tested macrolides, azithromycin, clarithromycin and roxithromycin, exhibited pronounced, concentration-related reduction of IL-1β, IL-6, IL-10, TNF-α, CCL3, CCL5, CCL20, CCL22, CXCL1, CXCL5, and G-CSF release. Further slight inhibitory effects on IL-1α, CXCL8, GM-CSF, and PAI-1 production were also observed. Erythromycin was very weakly active. Qualitatively and quantitatively, macrolides exerted distinctive and, compared to other tested classes of compounds, more pronounced immunomodulatory effects, particularly in terms of chemokine (CCL3, CCL5, CCL20, CCL22, and CXCL5), IL-1β, G-CSF and PAI-1 release. The described modulation of inflammatory mediators could potentially contribute to further definition of biomarkers of macrolide anti-inflammatory activity in COPD., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
42. Increased phasic activity of VTA dopamine neurons in mice 3 weeks after repeated social defeat.
- Author
-
Razzoli M, Andreoli M, Michielin F, Quarta D, and Sokal DM
- Subjects
- Action Potentials physiology, Animals, Electrophysiology, Hyperphagia physiopathology, Mice, Dominance-Subordination, Dopamine metabolism, Neurons physiology, Ventral Tegmental Area physiology
- Abstract
Social defeat is an ethologically relevant stress inducing neuroadaptive changes in the mesocorticolimbic dopaminergic system. Three weeks after 10 days of daily defeat salient behaviors and in vivo dopamine (DA) neuron firing were evaluated in mice. Prior defeat induced social avoidance and hyperphagia and increased ventral tegmental area (VTA) DA neuron bursting activity. These data extend previous studies and suggest that increased phasic DA neuron firing in the VTA could be considered amongst the features defining the lasting imprint of social defeat stress., (Copyright © 2010 Elsevier B.V. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
43. Strain-specific outcomes of repeated social defeat and chronic fluoxetine treatment in the mouse.
- Author
-
Razzoli M, Carboni L, Andreoli M, Michielin F, Ballottari A, and Arban R
- Subjects
- Animals, Antidepressive Agents, Second-Generation blood, Biomarkers blood, Fluoxetine blood, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Organ Size, Species Specificity, Stress, Physiological, Swimming, Antidepressive Agents, Second-Generation pharmacology, Behavior, Animal drug effects, Fluoxetine pharmacology, Social Behavior
- Abstract
Social stress is a risk factor for affective disorders in vulnerable individuals. Although the biological nature of stress susceptibility/resilience remains to be elucidated, genetic variation is considered amongst the principal contributors to brain disorders. Furthermore, genetic predisposition may be determinant for the therapeutic outcome, as proposed for antidepressant treatments. In the present studies we compared the inherently diverse genetic backgrounds of 2 mouse strains by assessing the efficacy of a chronic antidepressant treatment in a repeated social stress procedure. C57BL/6J and BalbC mice underwent 10-day social defeats followed by 28-day fluoxetine treatment (10 mg/kg/mL, p.o.). In C57BL/6J, most of the social defeat-induced changes were of metabolic nature including persistently altered feed efficiency and decreased abdominal fat stores that were ameliorated by fluoxetine. BalbC mouse behavior was persistently affected by social defeat both in the social avoidance and the forced swim tests, and in either procedure it was restored by chronic fluoxetine, whereas their endocrine parameters were mostly unaffected. The highlighted strain-specific responsivity to the metabolic and behavioral consequences of social defeat and to the chronic antidepressant treatment offers a promising research tool to further explore the underlying neural mechanisms and genetic basis of stress susceptibility and treatment response., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
44. GSK1614343, a novel ghrelin receptor antagonist, produces an unexpected increase of food intake and body weight in rodents and dogs.
- Author
-
Costantini VJ, Vicentini E, Sabbatini FM, Valerio E, Lepore S, Tessari M, Sartori M, Michielin F, Melotto S, Bifone A, Pich EM, and Corsi M
- Subjects
- Animals, Body Composition drug effects, Dogs, Dose-Response Relationship, Drug, Female, Ghrelin blood, Ghrelin pharmacology, Hypothalamus drug effects, Hypothalamus metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Rats, Rats, Sprague-Dawley, Receptors, Ghrelin genetics, Stimulation, Chemical, Transcriptome drug effects, Azabicyclo Compounds pharmacology, Body Weight drug effects, Eating drug effects, Hydrazines pharmacology, Receptors, Ghrelin antagonists & inhibitors
- Abstract
Ghrelin is a 28-amino-acid polypeptide expressed in the stomach and hypothalamus that stimulates GH secretion, increases food intake (FI) and promotes body weight (BW) gain most likely via activation of the growth hormone secretagogue receptor type 1a (GHSR1a). GSK1614343 is a novel selective and potent GHSR antagonist with no partial agonist properties, recently characterized as GH secretion inhibitor by Sabbatini et al. [Chem Med Chem 2010;5:1450-1455]. In the present study, GSK1614343 (10 mg/kg) was not able to antagonize ghrelin-induced food consumption in rat, but unexpectedly stimulated FI and BW gain in both rats and dogs, a profile associated with decreased ghrelin plasma level. Interestingly, GSK1614343 selectively reduced the pro-opiomelanocortin mRNA levels in rat hypothalami chronically treated with the compound. To better understand the observed effects, we administered GSK1614343 (30 mg/kg) to Ghsr null mice and measured body mass components (fat, lean and free fluid) by using a NMR spectrometer. The increases of FI and BW were abolished in Ghsr null mice, while fat and lean masses increased in wild-type mice. Taken together, these results indicate that the orexigenic effect of GSK1614343 is mediated by GHSR1a and that the weight gain could be attributed to the increase of both adiposity and muscle mass, but not to fluid retention. The observed dissociation between effects on GH secretion and effects on FI/BW is inconsistent with a simple hormone-receptor model, suggesting unknown underlying regulations of the ghrelin system whose understanding require further investigation., (Copyright © 2011 S. Karger AG, Basel.)
- Published
- 2011
- Full Text
- View/download PDF
45. Circadian profile of peripheral hormone levels in Sprague-Dawley rats and in common marmosets (Callithrix jacchus).
- Author
-
Bertani S, Carboni L, Criado A, Michielin F, Mangiarini L, and Vicentini E
- Subjects
- Animals, Brain-Derived Neurotrophic Factor blood, Callithrix, Corticosterone blood, Female, Hydrocortisone blood, Insulin-Like Growth Factor I metabolism, Male, Rats, Rats, Sprague-Dawley, Species Specificity, Circadian Rhythm, Cytokines blood, Peptide Hormones blood
- Abstract
Aim: in the present study, we report the circadian profiles of a wide panel of hormones measured in rats and common marmosets (Callithrix jacchus), under physiological conditions, paying special attention to minimising the stress imposed on the animals., Materials and Methods: blood collections were performed over a 24-hour period for the analysis of stress and pituitary hormones, metabolic markers and cytokines from male cannulated rats connected to a fully automatic system, and healthy marmosets in which gender differences were also evaluated., Results: in rats, a significant time effect was observed for corticosterone, prolactin (PRL), thyroid stimulating hormone (TSH), growth hormone, follicle-stimulating hormone, brain-derived neurotrophic factor, total ghrelin, insulin, leptin, insulin-like growth factor-1, adiponectin and interleukin-10. In marmosets, a significant time effect for cortisol, adrenocorticotropic hormone (ACTH), PRL and TSH, with gender effect for ACTH and PRL only, was observed. On the contrary, luteinizing hormone in the rat and active ghrelin, peptide YY, pancreatic polypeptide and gastric inhibitory polypeptide in the marmoset did not show any significant circadian variation., Conclusion: the present work confirmed that, due to time-of-day dependent modulation of hormones, circadian rhythmicity is relevant in physiological studies and should also be taken into consideration when performing pharmacological studies.
- Published
- 2010
46. [Epidemiology and prevention of rheumatic fever in Rio Grande do Sul].
- Author
-
Michielin F, Pretto AA, Prataviera JC, Holz JL, Paternoster R, Catalunha M, Michielin Filho F, and Michielin JH
- Subjects
- Brazil epidemiology, Humans, Male, Rheumatic Fever prevention & control, Risk Factors, Rheumatic Fever epidemiology
- Published
- 1994
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.