378 results on '"Bottazzi, A"'
Search Results
2. Immunomics-guided discovery of serum and urine antibodies for diagnosing urogenital schistosomiasis: a biomarker identification study
- Author
-
Cornelis H. Hokke, Donald P. McManus, Alex Loukas, Govert J. van Dam, Takafira Mduluza, Said M. Ali, Thewarach Laha, Al Jasinskas, Maria Elena Bottazzi, Mark S. Pearson, Beatrice Greco, Sujittra Chaiyadet, A.A. Adegnika, Paul L. A. M. Corstjens, Matthew A. Field, Abena S. Amoah, Denise L. Doolan, Gebeyaw G Mekonnen, Javier Sotillo, Fatma Kabole, Claude Oeuvray, Bemnet Amare Tedla, Carla Proietti, Philip L. Felgner, Stefanie Knopp, Rie Nakajima, Luke Becker, David Rollinson, Pengfei Cai, Francisca Mutapi, Australian Trade and Investment Commission, Fundación Merck Salud, Bill & Melinda Gates Foundation, Australian Institute of Tropical Health and Medicine, University of Georgia (Estados Unidos), National Health and Medical Research Council (Australia), James Cook University (Australia), Wellcome Trust, Thrasher Research Fund, and European and Developing Countries Clinical Trials
- Subjects
Microbiology (medical) ,Male ,Medicine (General) ,Proteome ,Schistosomiasis ,Urine ,Microbiology ,Immunomics ,Schistosomiasis haematobia ,R5-920 ,Antigen ,Virology ,parasitic diseases ,Medicine ,Animals ,Humans ,Schistosoma haematobium ,biology ,business.industry ,Area under the curve ,Australia ,Extracellular vesicle ,Articles ,biology.organism_classification ,medicine.disease ,QR1-502 ,Infectious Diseases ,Immunoglobulin G ,Immunology ,biology.protein ,Female ,Antibody ,business ,Biomarkers - Abstract
Background: Sensitive diagnostics are needed for effective management and surveillance of schistosomiasis so that current transmission interruption goals set by WHO can be achieved. We aimed to screen the Schistosoma haematobium secretome to find antibody biomarkers of schistosome infection, validate their diagnostic performance in samples from endemic populations, and evaluate their utility as point of care immunochromatographic tests (POC-ICTs) to diagnose urogenital schistosomiasis in the field. Methods: We did a biomarker identification study, in which we constructed a proteome array containing 992 validated and predicted proteins from S haematobium and screened it with serum and urine antibodies from endemic populations in Gabon, Tanzania, and Zimbabwe. Arrayed antigens that were IgG-reactive and a select group of antigens from the worm extracellular vesicle proteome, predicted to be diagnostically informative, were then evaluated by ELISA using the same samples used to probe arrays, and samples from individuals residing in a low-endemicity setting (ie, Pemba and Unguja islands, Zanzibar, Tanzania). The two most sensitive and specific antigens were incorporated into POC-ICTs to assess their ability to diagnose S haematobium infection from serum in a field-deployable format. Findings: From array probing, in individuals who were infected, 208 antigens were the targets of significantly elevated IgG responses in serum and 45 antigens were the targets of significantly elevated IgG responses in urine. Of the five proteins that were validated by ELISA, Sh-TSP-2 (area under the curve [AUC]serum=0·98 [95% CI 0·95-1·00]; AUCurine=0·96 [0·93-0·99]), and MS3_01370 (AUCserum=0·93 [0·89-0·97]; AUCurine=0·81 [0·72-0·89]) displayed the highest overall diagnostic performance in each biofluid and exceeded that of S haematobium-soluble egg antigen in urine (AUC=0·79 [0·69-0·90]). When incorporated into separate POC-ICTs, Sh-TSP-2 showed absolute specificity and a sensitivity of 75% and MS3_01370 showed absolute specificity and a sensitivity of 89%. Interpretation: We identified numerous biomarkers of urogenital schistosomiasis that could form the basis of novel antibody diagnostics for this disease. Two of these antigens, Sh-TSP-2 and MS3_01370, could be used as sensitive, specific, and field-deployable diagnostics to support schistosomiasis control and elimination initiatives, with particular focus on post-elimination surveillance. This study received financial support from Merck Global Health Institute and the Australian Trade and Investment Commission (Australian Tropical Medicine Commercialisation grants programme ATMC50322). Research for the Zanzibar Elimination of Schistosomiasis Transmission project was funded by the University of Georgia Research Foundation, which is funded by the Bill & Melinda Gates Foundation for the Schistosomiasis Consortium for Operational Research and Evaluation projects (prime award number 50816, sub-award number RR374-053/4893206). SK received financial support by sub-award number RR374-053/4893196 and via direct grants from the Gates Foundation (investment identification numbers OPP1191423 and OPP1198086). AL was funded by a National Health and Medical Research Council Senior Principal Research Fellowship (number APP1117504). BAT was funded by a James Cook University Postgraduate Scholarship. GGM was funded by an Australian Institute of Tropical Health and Medicine postgraduate scholarship. Funding was granted to AAA by a European and Developing Countries Clinical Trials Partnership senior fellowship training award (number TA_11_40200). FM was funded by the Thrasher Research Fund (number 12440) and the Wellcome Trust (number 108061/Z/15/Z). Sí
- Published
- 2021
- Full Text
- View/download PDF
3. SARS‑CoV-2 RBD219-N1C1: A yeast-expressed SARS-CoV-2 recombinant receptor-binding domain candidate vaccine stimulates virus neutralizing antibodies and T-cell immunity in mice
- Author
-
Brian Keegan, Jungsoon Lee, Rahki Kundu, Bin Zhan, Wen-Hsiang Chen, Zhuyun Liu, Portia M. Gillespie, Leroy Versteeg, Jeroen Pollet, Zoha Momin, Jason T. Kimata, Ana Carolina de Araujo Leao, Cristina Poveda, Maria Jose Villar, Rakesh Adhikari, Joanne Altieri Rivera, Maria Elena Bottazzi, Junfei Wei, Ulrich Strych, and Peter J. Hotez
- Subjects
COVID-19 Vaccines ,T-Lymphocytes ,030231 tropical medicine ,Immunology ,coronavirus ,Saccharomyces cerevisiae ,Antibodies, Viral ,medicine.disease_cause ,Article ,Virus ,RBD ,law.invention ,Pichia pastoris ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,Antigen ,law ,medicine ,Animals ,Humans ,Immunology and Allergy ,030212 general & internal medicine ,Neutralizing antibody ,Coronavirus ,Pharmacology ,Mice, Inbred BALB C ,pseudovirus ,biology ,SARS-CoV-2 ,Ligand binding assay ,Immunogenicity ,COVID-19 ,biology.organism_classification ,Antibodies, Neutralizing ,Virology ,alum ,Saccharomycetales ,Spike Glycoprotein, Coronavirus ,Recombinant DNA ,biology.protein ,Antibody ,ACE-2 ,Research Article - Abstract
There is an urgent need for an accessible and low-cost COVID-19 vaccine suitable for low- and middle-income countries. Here we report on the development of a SARS-CoV-2 receptor-binding domain (RBD) protein, expressed at high levels in yeast (Pichia pastoris), as a suitable vaccine candidate against COVID-19. After introducing two modifications into the wild-type RBD gene to reduce yeast-derived hyperglycosylation and improve stability during protein expression, we show that the recombinant protein, RBD219-N1C1, is equivalent to the wild-type RBD recombinant protein (RBD219-WT) in anin vitroACE-2 binding assay. Immunogenicity studies of RBD219-N1C1 and RBD219-WT proteins formulated with Alhydrogel®were conducted in mice, and, after two doses, both the RBD219-WT and RBD219-N1C1 vaccines induced high levels of binding IgG antibodies. Using a SARS-CoV-2 pseudovirus, we further showed that sera obtained after a two-dose immunization schedule of the vaccines were sufficient to elicit strong neutralizing antibody titers in the 1:1,000 to 1:10,000 range, for both antigens tested. The vaccines induced IFN-γ, IL-6, and IL-10 secretion, among other cytokines. Overall, these data suggest that the RBD219-N1C1 recombinant protein, produced in yeast, is suitable for further evaluation as a human COVID-19 vaccine, in particular, in an Alhydrogel®containing formulation and possibly in combination with other immunostimulants.
- Published
- 2021
- Full Text
- View/download PDF
4. Alterations to the Cardiac Metabolome Induced by Chronic T. cruzi Infection Relate to the Degree of Cardiac Pathology
- Author
-
Zongyuan Liu, Kristyn Hoffman, Kathryn M. Jones, Peter J. Hotez, Laura-Isobel McCall, Maria Elena Bottazzi, and Ekram Hossain
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,biology ,business.industry ,Cardiac fibrosis ,030106 microbiology ,Inflammation ,medicine.disease ,biology.organism_classification ,Pathogenesis ,03 medical and health sciences ,030104 developmental biology ,Infectious Diseases ,Fibrosis ,Pathognomonic ,cardiovascular system ,Metabolome ,Medicine ,Biomarker (medicine) ,medicine.symptom ,business ,Trypanosoma cruzi - Abstract
Chronic Chagasic cardiomyopathy (CCC) is a Neglected Tropical Disease caused by the parasite Trypanosoma cruzi. The pathognomonic findings in symptomatic CCC patients and animal models includes diffuse cardiac fibrosis and inflammation with persistent parasite presence in the heart. This study investigated chemical alterations in different regions of the heart in relation to cardiac pathology indicators to better understand the long-term pathogenesis of this neglected disease. We used data from echocardiography, fibrosis biomarkers, and histopathological analysis to fully evaluate cardiac pathology. Metabolites isolated from the pericardial and endocardial sides of the right ventricular myocardium were analyzed by liquid chromatography tandem mass spectrometry. The endocardial sections contained significantly less cardiac inflammation and fibrosis than the pericardial sections. Cardiac levels of acylcarnitines, phosphocholines, and other metabolites were significantly disrupted in accordance with cardiac fibrosis, inflammation, and serum fibrosis biomarker levels. These findings have potential implications in treatment and monitoring for CCC patients.
- Published
- 2021
- Full Text
- View/download PDF
5. Advances in vaccine development for human trichuriasis
- Author
-
Jesica Hayon, Jill Weatherhead, Peter J. Hotez, Maria Elena Bottazzi, and Bin Zhan
- Subjects
0301 basic medicine ,biology ,Trichuriasis ,Immunogenicity ,030231 tropical medicine ,Helminthiasis ,medicine.disease ,biology.organism_classification ,03 medical and health sciences ,Malnutrition ,030104 developmental biology ,0302 clinical medicine ,Infectious Diseases ,parasitic diseases ,Immunology ,medicine ,Helminths ,Trichuris trichiura ,Animal Science and Zoology ,Parasitology ,Ascaris lumbricoides ,Mass drug administration - Abstract
Trichuriasis known as whipworm infection caused by Trichuris trichiura, is a highly prevalent soil-transmitted helminthiasis in low- and middle-income countries located in tropical and subtropical areas and affecting approximately 360 million people. Children typically harbour the largest burden of T. trichiura and they are usually co-infected with other soil-transmitted helminth (STH), including Ascaris lumbricoides and hookworm. The consequences of trichuriasis, such as malnutrition and physical and cognitive growth restriction, lead to a massive health burden in endemic regions. Despite the implementation of mass drug administration of anthelminthic treatment to school-age children, T. trichiura infection remains challenging to control due to the low efficacy of current drugs as well as high rates of post-treatment re-infection. Thus, the development of a vaccine that would induce protective immunity and reduce infection rate or community faecal egg output is essential. Hurdles for human whipworm vaccine development include the lack of suitable vaccine antigen targets and animal models for human T. trichiura infection. Instead, rodent whipworm T. muris infected mouse models serve as a major surrogate for testing immunogenicity and efficacy of vaccine candidates. In this review, we summarize recent advances in animal models for T. trichiura antigen discovery and testing of vaccine candidates, while providing an overall view of the current status of T. trichiura vaccine development.
- Published
- 2021
- Full Text
- View/download PDF
6. The interferon landscape along the respiratory tract impacts the severity of COVID-19
- Author
-
Riccardo Colombo, Ivan Zanoni, Andreas Wack, Achille Broggi, Nicola Clementi, Tommaso Fossali, Laura Marongiu, Elena Tagliabue, Andrea Bottazzi, Fabio A. Facchini, Sofia Sisti, Janet Chou, Roberto Spreafico, Roberto Ferrarese, Elena Criscuolo, Vanessa Frangipane, Jaclyn M. Long, Federica Meloni, Nicasio Mancini, Sara Bozzini, Stefania Crotta, Benedetta Sposito, Massimo Clementi, Enju Liu, Antonio E. Pontiroli, Laura Pandolfi, Alessandro Ambrosi, Laura Saracino, Harvard Medical School [Boston] (HMS), Centre d'Immunologie de Marseille - Luminy (CIML), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Università degli Studi di Pavia = University of Pavia (UNIPV), The Francis Crick Institute [London], Universita Vita Salute San Raffaele = Vita-Salute San Raffaele University [Milan, Italie] (UniSR), Institute for Quantitative and Computational Biosciences - University of california LA, Division of Gastroenterology [Boston, MA, USA], Harvard University-Harvard Medical School [Boston] (HMS)-Beth Israel Deaconess Medical Center [Boston] (BIDMC), Dipartimento di Informatica, Matematica, Elettronica e Trasporti [Reggio Calabria] (DIMET), Universita Mediterranea of Reggio Calabria [Reggio Calabria], Luigi sacco hospital - Division of Anesthesiology and intensive Care, IRCCS Multimedica, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, Università degli Studi di Milano = University of Milan (UNIMI), Division of Respiratory Diseases (IRCCS), Università degli Studi di Pavia = University of Pavia (UNIPV)-San Matteo' Hospital, IRCCS laboratory of medical microbiology and virology Milan, Harvard University [Cambridge]-Harvard Medical School [Boston] (HMS)-Beth Israel Deaconess Medical Center [Boston] (BIDMC), DUMENIL, Anita, Sposito, B, Broggi, A, Pandolfi, L, Crotta, S, Clementi, N, Ferrarese, R, Sisti, S, Criscuolo, E, Spreafico, R, Long, J, Ambrosi, A, Liu, E, Frangipane, V, Saracino, L, Bozzini, S, Marongiu, L, Facchini, F, Bottazzi, A, Fossali, T, Colombo, R, Clementi, M, Tagliabue, E, Chou, J, Pontiroli, A, Meloni, F, Wack, A, Mancini, N, Zanoni, I, Sposito, Benedetta, Broggi, Achille, Pandolfi, Laura, Crotta, Stefania, Clementi, Nicola, Ferrarese, Roberto, Sisti, Sofia, Criscuolo, Elena, Spreafico, Roberto, Long, Jaclyn M., Ambrosi, Alessandro, Liu, Enju, Frangipane, Vanessa, Saracino, Laura, Bozzini, Sara, Marongiu, Laura, Facchini, Fabio A., Bottazzi, Andrea, Fossali, Tommaso, Colombo, Riccardo, Clementi, Massimo, Tagliabue, Elena, Chou, Janet, Pontiroli, Antonio E., Meloni, Federica, Wack, Andrea, Mancini, Nicasio, and Zanoni, Ivan
- Subjects
Model organisms ,Aging ,dendritic cell ,pattern recognition receptor ,[SDV]Life Sciences [q-bio] ,Respiratory System ,Immunology ,Type I IFN ,Infectious Disease ,Biology ,Severity of Illness Index ,Article ,General Biochemistry, Genetics and Molecular Biology ,lung ,Interferon ,Type III IFN ,Severity of illness ,Leukocytes ,medicine ,Humans ,Respiratory system ,ComputingMilieux_MISCELLANEOUS ,Regulation of gene expression ,Respiratory Distress Syndrome ,Human Biology & Physiology ,Lung ,SARS-CoV-2 ,epithelial cell ,FOS: Clinical medicine ,Age Factors ,Pattern recognition receptor ,COVID-19 ,Epithelial Cells ,interferon ,Viral Load ,COVID-19, SARS-CoV-2, Type I IFN, Type III IFN, airways, dendritic cell, epithelial cell, interferon, lung ,[SDV] Life Sciences [q-bio] ,medicine.anatomical_structure ,Gene Expression Regulation ,airways ,airway ,Interferons ,Viral load ,Respiratory tract ,medicine.drug - Abstract
Severe COVID-19 is characterized by overproduction of immune mediators, but the role of interferons (IFNs) of the type I (IFN-I) or type III (IFN-III) families remains debated. We scrutinized the production of IFNs along the respiratory tract of COVID-19 patients and found that high levels of IFN-III, and to a lesser extent IFN-I, characterize the upper airways of patients with high viral burden but reduced disease risk or severity. Production of specific IFN-III, but not IFN-I, members, denotes patients with a mild pathology and efficiently drives the transcription of genes that protect against SARS-CoV-2. In contrast, compared to subjects with other infectious or non-infectious lung pathologies, IFNs are over-represented in the lower airways of patients with severe COVID-19 that exhibit gene pathways associated with increased apoptosis and decreased proliferation. Our data demonstrate a dynamic production of IFNs in SARS-CoV-2-infected patients and show IFNs play opposing roles at distinct anatomical sites., An in-depth analysis of interferons in COVID-19 reveals differences in their roles based on anatomical location, viral load, age and disease severity. In the upper respiratory tract, high levels of IFN-III are protective and result in mild disease in spite of higher SARS-CoV-2 viral burden while the lower airways of patients with severe COVID-19 demonstrate elevated IFN-I and III, cell death and a reduction in interferon stimulated genes.
- Published
- 2021
- Full Text
- View/download PDF
7. Vaccination with chimeric protein induces protection in murine model against ascariasis
- Author
-
Maria Elena Bottazzi, Joseane Camilla de Castro, Bin Zhan, Denise Silva Nogueira, Fabrício Marcus Silva Oliveira, Daniella Castanheira Bartholomeu, Laila Viana de Almeida, Lilian Lacerda Bueno, João Luís Reis-Cunha, Peter J. Hotez, Ricardo Toshio Fujiwara, Luísa Mourão Dias Magalhães, and Mariana Santos Cardoso
- Subjects
Recombinant Fusion Proteins ,030231 tropical medicine ,Monophosphoryl Lipid A ,Epitope ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,medicine ,Animals ,030212 general & internal medicine ,Ascaris suum ,B cell ,Ascariasis ,Mice, Inbred BALB C ,General Veterinary ,General Immunology and Microbiology ,biology ,Ascaris ,Vaccination ,Public Health, Environmental and Occupational Health ,biology.organism_classification ,Fusion protein ,Virology ,Disease Models, Animal ,Infectious Diseases ,medicine.anatomical_structure ,Antigens, Helminth ,Molecular Medicine ,Female - Abstract
An estimated 400 million people are infected by parasites of the genus Ascaris and the existing control measures are inefficient. Vaccine development using B cell antigens is a promising strategy for increased protection against this parasite. The present study aimed at developing a chimeric protein capable of conferring protection against infection by Ascaris sp. For this purpose, we performed B-cell epitope predictions on previously described vaccine candidate proteins from Ascaris suum and the corresponding peptides were used to construct a chimeric protein. Female BALB / c mice were immunized subcutaneously in three doses at 10 day intervals with a vaccine formulation comprised of the chimeric protein together with monophosphoryl lipid A (MPLA). Control groups included protein alone, MPLA, or PBS. After challenge infection, animals vaccinated with chimeric protein plus MPLA showed a reduction of 73.54% of larval load in the lung compared to control group animals. Animals immunized with chimeric protein plus MPLA also display higher IgG response and a reduction in lung inflammation. Our study highlights how chimeric proteins containing more than one B cell epitope can enhance immune protection against helminthic infection and offer new approaches to the development of Ascaris vaccines.
- Published
- 2021
- Full Text
- View/download PDF
8. Yeast-expressed SARS-CoV recombinant receptor-binding domain (RBD219-N1) formulated with aluminum hydroxide induces protective immunity and reduces immune enhancement
- Author
-
Ulrich Strych, Peter J. Hotez, Anurodh S. Agrawal, Jeroen Pollet, Lanying Du, Shibo Jiang, Wen-Hsiang Chen, Chien-Te K Tseng, Xinrong Tao, Bi Hung Peng, Abdullah Algaissi, Maria Elena Bottazzi, and Sara Lustigman
- Subjects
viruses ,medicine.medical_treatment ,coronavirus ,Aluminum Hydroxide ,Antibodies, Viral ,medicine.disease_cause ,law.invention ,Mice ,0302 clinical medicine ,law ,vaccine ,030212 general & internal medicine ,skin and connective tissue diseases ,Neutralizing antibody ,Coronavirus ,Mice, Inbred BALB C ,Vaccines, Synthetic ,biology ,Viral Vaccine ,Viral Load ,eosinophil infiltration ,Recombinant Proteins ,Infectious Diseases ,Spike Glycoprotein, Coronavirus ,Vaccines, Subunit ,Recombinant DNA ,Molecular Medicine ,Female ,Antibody ,Coronavirus Infections ,Viral load ,Adjuvant ,COVID-19 Vaccines ,Pneumonia, Viral ,030231 tropical medicine ,severe acute respiratory syndrome ,Article ,Virus ,Betacoronavirus ,03 medical and health sciences ,Immune system ,Protein Domains ,medicine ,Animals ,Humans ,Pandemics ,General Veterinary ,General Immunology and Microbiology ,SARS-CoV-2 ,fungi ,Public Health, Environmental and Occupational Health ,COVID-19 ,Viral Vaccines ,Antibodies, Neutralizing ,Virology ,respiratory tract diseases ,body regions ,biology.protein ,recombinant protein - Abstract
Highlights • A SARS-CoV recombinant protein-based receptor binding domain (RBD) vaccine on alum provides high neutralizing antibody titers eliciting 100% protection (survival) against homologous SARS CoV virus challenge. • A SARS-CoV RBD vaccine on alum prevents pulmonary cellular infiltrates upon virus challenge. • A SARS-CoV RBD vaccine on alum greatly reduces lung eosinophils compared to a vaccine comprised of the SARS-CoV S protein. • The SARS-CoV RBD vaccine on alum is being developed as a human vaccine., We developed a severe acute respiratory syndrome (SARS) subunit recombinant protein vaccine candidate based on a high-yielding, yeast- engineered, receptor-binding domain (RBD219-N1) of the SARS beta-coronavirus (SARS-CoV) spike (S) protein. When formulated with Alhydrogel®, RBD219-N1 induced high-level neutralizing antibodies against both pseudotyped virus and a clinical (mouse-adapted) isolate of SARS-CoV. Here, we report that mice immunized with RBD219-N1/Alhydrogel® were fully protected from lethal SARS-CoV challenge (0% mortality), compared to ∼ 30% mortality in mice immunized with the SARS S protein formulated with Alhydrogel®, and 100% mortality in negative controls. An RBD219-N1 formulation with Alhydrogel® was also superior to the S protein, unadjuvanted RBD, and AddaVax (MF59-like adjuvant)-formulated RBD in inducing specific antibodies and preventing cellular infiltrates in the lungs upon SARS-CoV challenge. Specifically, a formulation with a 1:25 ratio of RBD219-N1 to Alhydrogel® provided high neutralizing antibody titers, 100% protection with non-detectable viral loads with minimal or no eosinophilic pulmonary infiltrates. As a result, this vaccine formulation is under consideration for further development against SARS-CoV and potentially other emerging and re-emerging beta-CoVs such as SARS-CoV-2.
- Published
- 2020
- Full Text
- View/download PDF
9. Process Characterization and Biophysical Analysis for a Yeast-Expressed Phlebotomus papatasi Salivary Protein (PpSP15) as a Leishmania Vaccine Candidate
- Author
-
Rakhi Tyagi Kundu, Wen-Hsiang Chen, Mun Peak Nyon, Amadeo B. Biter, Peter J. Hotez, Ulrich Strych, Maria Elena Bottazzi, Mohan Vivekanandan Poongavanam, and Zhuyun Liu
- Subjects
Pharmaceutical Science ,Saccharomyces cerevisiae ,02 engineering and technology ,030226 pharmacology & pharmacy ,Microbiology ,law.invention ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cutaneous leishmaniasis ,law ,medicine ,Animals ,Parasite hosting ,Leishmania major ,Salivary Proteins and Peptides ,Leishmaniasis Vaccines ,biology ,Tropical disease ,021001 nanoscience & nanotechnology ,medicine.disease ,biology.organism_classification ,Leishmania ,Yeast ,Sandfly ,Phlebotomus ,Recombinant DNA ,Insect Proteins ,0210 nano-technology - Abstract
Cutaneous leishmaniasis is a neglected tropical disease caused by the parasite Leishmania and transmitted by sandflies. It has become a major health problem in many tropical and subtropical countries, especially in regions of conflict and political instability. Currently, there are only limited drug treatments and no available licensed vaccine; thus, the need for more therapeutic interventions remains urgent. Previously, a DNA vaccine encoding a 15 kDa sandfly (Phlebotomus papatasi) salivary protein (PpSP15) and recombinant nonpathogenic Leishmania tarentolae secreting PpSP15 have been shown to induce protective immunity against Leishmania major in mice, demonstrating that PpSP15 is a promising vaccine candidate. In this study, we developed a fermentation process in yeast with a yield of ~1g PpSP15/L and a scalable purification process consisting of only 2 chromatographic purification steps with high binding capacity for PpSP15, suggesting that PpSP15 can be produced economically. The biophysical/biochemical analysis of the purified PpSP15 indicated that the protein was of high purity (>97%) and conformationally stable between pH 4.4 and 9.0. More importantly, the recombinant protein had a defined structure similar to that of the related PdSP15 from Phlebotomus duboscqi, implying the suitability of the yeast expression system for producing a correctly folded PpSP15.
- Published
- 2020
- Full Text
- View/download PDF
10. The complement system inAspergillus fumigatusinfections and its crosstalk with pentraxins
- Author
-
Cecilia Garlanda, Barbara Bottazzi, Antonio Inforzato, Raffaella Parente, and Andrea Doni
- Subjects
Biophysics ,Biochemistry ,Aspergillus fumigatus ,03 medical and health sciences ,Immune system ,Structural Biology ,Genetics ,Animals ,Aspergillosis ,Humans ,Host-pathogen interface ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Innate immune system ,Pentraxins ,biology ,Effector ,030302 biochemistry & molecular biology ,Complement System Proteins ,Cell Biology ,biology.organism_classification ,Complement system ,Serum Amyloid P-Component ,Crosstalk (biology) ,C-Reactive Protein ,Immunology ,biology.protein - Abstract
Aspergillosis is a life-threatening infection mostly affecting immunocompromised individuals and primarily caused by the saprophytic fungus Aspergillus fumigatus. At the host-pathogen interface, both cellular and humoral components of the innate immune system are increasingly acknowledged as essential players in the recognition and disposal of this opportunistic mold. Fundamental hereof is the contribution of the complement system, which deploys all three activation pathways in the battle against A. fumigatus, and functionally cooperates with other soluble pattern recognition molecules, including pentraxins. In particular, preclinical and clinical observations point to the long pentraxin PTX3 as a nonredundant and complement-dependent effector with protective functions against A. fumigatus. Based on past and current literature, here we discuss how the complement participates in the immune response to this fungal pathogen, and illustrate its crosstalk with the pentraxins, with a focus on PTX3. Emphasis is placed on the molecular mechanisms underlying such processes, the genetic evidence from human epidemiology, and the translational potential of the currently available knowledge.
- Published
- 2020
- Full Text
- View/download PDF
11. 3D Cocultures of Osteoblasts and Staphylococcus aureus on Biomimetic Bone Scaffolds as a Tool to Investigate the Host–Pathogen Interface in Osteomyelitis
- Author
-
Monica Sandri, Anna Tampieri, Barbara Bottazzi, Ciro Menale, Valentina Possetti, Antonio Inforzato, Mattia Loppini, Maria Lucia Schiavone, Andrea Doni, Raffaella Parente, Elisabetta Campodoni, Alberto Mantovani, Cristina Sobacchi, Parente, R., Possetti, V., Schiavone, M. L., Campodoni, E., Menale, C., Loppini, M., Doni, A., Bottazzi, B., Mantovani, A., Sandri, M., Tampieri, A., Sobacchi, C., and Inforzato, A.
- Subjects
3D model ,0301 basic medicine ,Microbiology (medical) ,Staphylococcus aureus ,030106 microbiology ,3d model ,Biology ,medicine.disease_cause ,Article ,Extracellular matrix ,03 medical and health sciences ,Immune system ,Biomimetic bone scaffold ,Gene expression ,Osteomyeliti ,medicine ,Immunology and Allergy ,Host-pathogen interface ,osteoblast-like cells ,Molecular Biology ,General Immunology and Microbiology ,Osteoblast-like cell ,osteomyelitis ,host-pathogen interface ,3D models ,biomimetic bone scaffolds ,Osteomyelitis ,medicine.disease ,In vitro ,Cell biology ,030104 developmental biology ,Infectious Diseases ,Medicine ,host–pathogen interface - Abstract
Osteomyelitis (OM) is an infectious disease of the bone primarily caused by the opportunistic pathogen Staphylococcus aureus (SA). This Gram-positive bacterium has evolved a number of strategies to evade the immune response and subvert bone homeostasis, yet the underlying mechanisms remain poorly understood. OM has been modeled in vitro to challenge pathogenetic hypotheses in controlled conditions, thus providing guidance and support to animal experimentation. In this regard, traditional 2D models of OM inherently lack the spatial complexity of bone architecture. Three-dimensional models of the disease overcome this limitation, however, they poorly reproduce composition and texture of the natural bone. Here, we developed a new 3D model of OM based on cocultures of SA and murine osteoblastic MC3T3-E1 cells on magnesium-doped hydroxyapatite/collagen I (MgHA/Col) scaffolds that closely recapitulate the bone extracellular matrix. In this model, matrix-dependent effects were observed in proliferation, gene transcription, protein expression, and cell–matrix interactions both of the osteoblastic cell line and of bacterium. Additionally, these had distinct metabolic and gene expression profiles, compared to conventional 2D settings, when grown on MgHA/Col scaffolds in separate monocultures. Our study points to MgHA/Col scaffolds as biocompatible and bioactive matrices and provides a novel and close-to-physiology tool to address the pathogenetic mechanisms of OM at the host–pathogen interface.
- Published
- 2021
- Full Text
- View/download PDF
12. Complementary Roles of Short and Long Pentraxins in the Complement-Mediated Immune Response to Aspergillus fumigatus Infections
- Author
-
Raffaella Parente, Valentina Possetti, Marco Erreni, Francesca D’Autilia, Barbara Bottazzi, Cecilia Garlanda, Alberto Mantovani, Antonio Inforzato, and Andrea Doni
- Subjects
Mini Review ,Immunology ,Biology ,Microbiology ,Aspergillus fumigatus ,Immune system ,Immunology and Allergy ,Animals ,Humans ,aspergillosis ,complement ,Opsonin ,Complement Activation ,innate immunity ,pentraxins ,Innate immune system ,Pentraxins ,Pattern recognition receptor ,PTX3 ,Complement System Proteins ,RC581-607 ,biology.organism_classification ,Immunity, Innate ,Complement system ,Serum Amyloid P-Component ,C-Reactive Protein ,Host-Pathogen Interactions ,biology.protein ,Disease Susceptibility ,Immunologic diseases. Allergy ,Biomarkers - Abstract
The ubiquitous moldAspergillus fumigatusis the major etiologic agent of invasive aspergillosis, a life-threatening infection amongst immune compromised individuals. An increasing body of evidence indicates that effective disposal ofA. fumigatusrequires the coordinate action of both cellular and humoral components of the innate immune system. Early recognition of the fungal pathogen, in particular, is mediated by a set of diverse soluble pattern recognition molecules (PRMs) that act as “ancestral antibodies” inasmuch as they are endowed with opsonic, pro-phagocytic and killing properties. Pivotal is, in this respect, the contribution of the complement system, which functionally cooperates with cell-borne pattern recognition receptors (PRRs) and other soluble PRMs, including pentraxins. Indeed, complement and pentraxins form an integrated system with crosstalk, synergism, and regulation, which stands as a paradigm of the interplay between PRMs in the mounting and orchestration of antifungal immunity. Following upon our past experience with the long pentraxin PTX3, a well-established immune effector in the host response toA. fumigatus, we recently reported that this fungal pathogen is targetedin vitroandin vivoby the short pentraxin Serum Amyloid P component (SAP) too. Similar to PTX3, SAP promotes phagocytosis and disposal of the fungal pathogenviacomplement-dependent pathways. However, the two proteins exploit different mechanisms of complement activation and receptor-mediated phagocytosis, which further extends complexity and integration of the complement-pentraxin crosstalk in the immune response toA. fumigatus. Here we revisit this crosstalk in light of the emerging roles of SAP as a novel PRM with antifungal activity.
- Published
- 2021
- Full Text
- View/download PDF
13. Monocyte-macrophage polarization and recruitment pathways in the tumour microenvironment of B-cell acute lymphoblastic leukaemia
- Author
-
Alessandra Fallati, Oscar Maglia, Giovanna D'Amico, Fabio Pagni, Mariella D'Angiò, Erica Dander, Federica Portale, Giulia Cricrì, Barbara Bottazzi, Fabio Pasqualini, Rita Starace, Lisa Brizzolara, A Mantovani, Chiara Buracchi, Paola Allavena, Gloria Bedini, Maria Grazia Valsecchi, Stefania Gaspari, Franco Locatelli, Andrea Biondi, Tamara Gulic, Daniela Silvestri, Cecilia Garlanda, Dander, E, Fallati, A, Gulic, T, Pagni, F, Gaspari, S, Silvestri, D, Cricri, G, Bedini, G, Portale, F, Buracchi, C, Starace, R, Pasqualini, F, D'Angio, M, Brizzolara, L, Maglia, O, Mantovani, A, Garlanda, C, Valsecchi, M, Locatelli, F, Biondi, A, Bottazzi, B, Allavena, P, and D'Amico, G
- Subjects
Adult ,Male ,Chemokine ,acute lymphoblastic leukaemia ,Adolescent ,CD14 ,chemokines ,macrophage ,Monocytes ,03 medical and health sciences ,0302 clinical medicine ,Antigens, CD ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,medicine ,Human Umbilical Vein Endothelial Cells ,Tumor Microenvironment ,Macrophage ,Humans ,CX3CL1 ,Aged ,bone marrow microenvironment ,biology ,Chemistry ,Monocyte ,Macrophages ,Mesenchymal stem cell ,chemokine ,Hematology ,Middle Aged ,Coculture Techniques ,Neoplasm Proteins ,medicine.anatomical_structure ,Settore MED/38 - PEDIATRIA GENERALE E SPECIALISTICA ,030220 oncology & carcinogenesis ,mesenchymal stromal cell ,monocyte ,Cancer research ,biology.protein ,Female ,Bone marrow ,mesenchymal stromal cells ,CD163 ,030215 immunology - Abstract
B-cell acute lymphoblastic leukaemia (B-ALL) reprograms the surrounding bone marrow (BM) stroma to create a leukaemia-supportive niche. To elucidate the contribution of immune cells to the leukaemic microenvironment, we investigated the involvement of monocyte/macrophage compartments, as well as several recruitment pathways in B-ALL development. Immunohistochemistry analyses showed that CD68-expressing macrophages were increased in leukaemic BM biopsies, compared to controls and predominantly expressed the M2-like markers CD163 and CD206. Furthermore, the "non-classical" CD14+ CD16++ monocyte subset, expressing high CX3CR1 levels, was significantly increased in B-ALL patients' peripheral blood. CX3CL1 was shown to be significantly upregulated in leukaemic BM plasma, thus providing an altered migratory pathway possibly guiding NC monocyte recruitment into the BM. Additionally, the monocyte/macrophage chemoattractant chemokine ligand 2 (CCL2) strongly increased in leukaemic BM plasma, possibly because of the interaction of leukaemic cells with mesenchymal stromal cells and vascular cells and due to a stimulatory effect of leukaemia-related inflammatory mediators. C5a, a macrophage chemoattractant and M2-polarizing factor, further appeared to be upregulated in the leukaemic BM, possibly as an effect of PTX3 decrease, that could unleash complement cascade activation. Overall, deregulated monocyte/macrophage compartments are part of the extensive BM microenvironment remodelling at B-ALL diagnosis and could represent valuable targets for novel treatments to be coupled with classical chemotherapy.
- Published
- 2020
14. Protective efficacy in a hamster model of a multivalent vaccine for human visceral leishmaniasis (Mulevaclin) consisting of the kmp11, leish-f3+, and ljl143 antigens in virosomes, plus gla-se adjuvant
- Author
-
Anabela Cordeiro-da-Silva, Jesus G. Valenzuela, Laura Fernández, Mª Ángeles Jiménez, Epifanio Fichera, Gaurav Gupta, Begoña Pérez-Cabezas, Maria Elena Bottazzi, Carmen Sánchez, Reinhard Glueck, Pedro Cecílio, Eugenia Carrillo, Fabiano Oliveira, Rhea N. Coler, Reed Steven G, Shaden Kamhawi, Javier Moreno, Luigi Gradoni, José Carlos Solana, Jose M. Requena, European Commission, Red de Investigación Cooperativa en Enfermedades Tropicales (España), Unión Europea. Comisión Europea. 7 Programa Marco, Plan Nacional de I+D+i (España), and European Regional Development Fund
- Subjects
Microbiology (medical) ,GLA-SE ,Virosomes ,QH301-705.5 ,medicine.medical_treatment ,macromolecular substances ,Microbiology ,Article ,Immune system ,Antigen ,Virology ,KMP11 ,LJL143 ,medicine ,Hamster ,Biology (General) ,Leishmaniasis ,biology ,LEISH-F3 ,business.industry ,biology.organism_classification ,medicine.disease ,Leishmania ,Vaccination ,Visceral leishmaniasis ,Leishmania infantum ,business ,Adjuvant ,Vaccine - Abstract
Visceral leishmaniasis (VL) is the most severe clinical form of leishmaniasis, fatal if untreated. Vaccination is the most cost-effective approach to disease control; however, to date, no vaccines against human VL have been made available. This work examines the efficacy of a novel vaccine consisting of the Leishmania membrane protein KMP11, LEISH-F3+ (a recombinant fusion protein, composed of epitopes of the parasite proteins nucleoside hydrolase, sterol-24-c-methyltransferase, and cysteine protease B), and the sand fly salivary protein LJL143, in two dose ratios. The inclusion of the TLR4 agonist GLA-SE as an adjuvant, and the use of virosomes (VS) as a delivery system, are also examined. In a hamster model of VL, the vaccine elicited antigen-specific immune responses prior to infection with Leishmania infantum. Of note, the responses were greater when higher doses of KMP11 and LEISH-F3+ proteins were administered along with the GLA-SE adjuvant and/or when delivered within VS. Remarkably, hamsters immunized with the complete combination (i.e., all antigens in VS + GLA-SE) showed significantly lower parasite burdens in the spleen compared to those in control animals. This protection was underpinned by a more intense, specific humoral response against the KMP11, LEISH-F3+, and LJL143 antigens in vaccinated animals, but a significantly less intense antibody response to the pool of soluble Leishmania antigens (SLA). Overall, these results indicate that this innovative vaccine formulation confers protection against L. infantum infection, supporting the advancement of the vaccine formulation into process development and manufacturing and the conduction of toxicity studies towards future phase I human clinical trials, European Community’s Seventh Framework Programme, grant number 603181 (Clinical Studies on a Multivalent Vaccine for Human Visceral Leishmaniasis [MuLeVaClin]), and by the RD16CIII/0003/0002 and RD16/0027/0008 Red de Investigación Cooperativa de Enfermedades Tropicales, Subprograma RETICS del Plan Estatal de I+D+I 2013–2016, co-funded by ERDF
- Published
- 2021
15. Characterization of T cell responses to co-administered hookworm vaccine candidates Na-GST-1 and Na-APR-1 in healthy adults in Gabon
- Author
-
Frejus Jeannot Zinsou, David Diemert, Jeffrey M. Bethony, Simon P. Jochems, Martin P. Grobusch, Yabo Josiane Honkpehedji, Sophie De Vries, Maria Elena Bottazzi, Peter J. Hotez, Remko van Leeuwen, Peter G. Kremsner, Friederike Sonnet, Jean-Claude Dejon Agobe, Ayola A. Adegnika, Yoanne D. Mouwenda, Koen A. Stam, Lucja A. Labuda, Marguerite Massinga Loembe, Madeleine E. Betouke Ongwe, Maria Yazdanbakhsh, APH - Global Health, AII - Infectious diseases, Graduate School, Infectious diseases, and APH - Aging & Later Life
- Subjects
Ancylostomatoidea ,Male ,Physiology ,T-Lymphocytes ,medicine.medical_treatment ,RC955-962 ,Biochemistry ,White Blood Cells ,Medical Conditions ,Animal Cells ,Arctic medicine. Tropical medicine ,Immune Physiology ,Medicine and Health Sciences ,CTLA-4 Antigen ,Public and Occupational Health ,Immunity, Cellular ,Vaccines ,Innate Immune System ,Immune System Proteins ,biology ,T Cells ,Vaccination ,Eukaryota ,Middle Aged ,Vaccination and Immunization ,Infectious Diseases ,Cytokine ,medicine.anatomical_structure ,Helminth Infections ,Cytokines ,Female ,Public aspects of medicine ,RA1-1270 ,Cellular Types ,Antibody ,Adjuvant ,Research Article ,medicine.drug ,Adult ,Infectious Disease Control ,Immune Cells ,T cell ,Immunology ,Antibodies, Helminth ,Peripheral blood mononuclear cell ,Antibodies ,Hookworm Infections ,Young Adult ,Adjuvants, Immunologic ,Antigen ,Helminths ,Vaccine Development ,Parasitic Diseases ,medicine ,Animals ,Humans ,Gabon ,Hookworm vaccine ,Blood Cells ,business.industry ,Organisms ,Public Health, Environmental and Occupational Health ,Biology and Life Sciences ,Proteins ,Cell Biology ,Molecular Development ,Invertebrates ,Hookworms ,Antigens, Helminth ,Immune System ,Antibody Formation ,biology.protein ,Preventive Medicine ,business ,Zoology ,Developmental Biology - Abstract
Two hookworm vaccine candidates, Na-GST-1 and Na-APR-1, formulated with Glucopyranosyl Lipid A (GLA-AF) adjuvant, have been shown to be safe, well tolerated, and to induce antibody responses in a Phase 1 clinical trial (Clinicaltrials.gov NCT02126462) conducted in Gabon. Here, we characterized T cell responses in 24 Gabonese volunteers randomized to get vaccinated three times with Na-GST-1 and Na-APR-1 at doses of 30μg (n = 8) or 100μg (n = 10) and as control Hepatitis B (n = 6). Blood was collected pre- and post-vaccination on days 0, 28, and 180 as well as 2-weeks after each vaccine dose on days 14, 42, and 194 for PBMCs isolation. PBMCs were stimulated with recombinant Na-GST-1 or Na-APR-1, before (days 0, 28 and 180) and two weeks after (days 14, 42 and 194) each vaccination and used to characterize T cell responses by flow and mass cytometry. A significant increase in Na-GST-1 -specific CD4+ T cells producing IL-2 and TNF, correlated with specific IgG antibody levels, after the third vaccination (day 194) was observed. In contrast, no increase in Na-APR-1 specific T cell responses were induced by the vaccine. Mass cytometry revealed that, Na-GST-1 cytokine producing CD4+ T cells were CD161+ memory cells expressing CTLA-4 and CD40-L. Blocking CTLA-4 enhanced the cytokine response to Na-GST-1. In Gabonese volunteers, hookworm vaccine candidate, Na-GST-1, induces detectable CD4+ T cell responses that correlate with specific antibody levels. As these CD4+ T cells express CTLA-4, and blocking this inhibitory molecules resulted in enhanced cytokine production, the question arises whether this pathway can be targeted to enhance vaccine immunogenicity., Author summary Two hookworm vaccine candidate (Na-GST-1 and Na-APR-1) have been tested in Gabonese and found to be safe and to induce antibody response. We aimed to study the cellular immune responses among vaccinated and unvaccinated volunteers. We found that Na-GST-1 induced CD4+ T cell responses (IL-2, TNF) among the vaccinated volunteers that received the high vaccine dose (100 ug). Furthermore Na-GST-1 specific memory T cells were found to express the inhibitory molecule CTLA-4. These responses was not observed in those who received the low dose of the Na-GST-1 vaccine, or those who received Na-APR-1 or HBV. By blocking CTLA-4, we observed an increase in TNF production. Our data suggest that an intervention involving blockage of the CTLA-4 molecule in the vaccinated could be beneficial in endemic settings where vaccine responses have been shown to be lower compared to non-endemic settings.
- Published
- 2021
16. Identification of vaccine targets in pathogens and design of a vaccine using computational approaches
- Author
-
Prashant Singh, P Preeti, Amit Chaudhary, Preeti Subramani, Swarsat Kaushik Nath, Ulrich Strych, Peter J. Hotez, Trapti Sharma, Maria Elena Bottazzi, Astha Mishra, Pranjay Gupta, Priya Kumari, Aman Kumar Dubey, Srijanee Gupta, Shriya Sood, Bilal Ahmed Abbasi, Kamal Rawal, Robin Sinha, Shachee Singh, Devansh Saraf, and Kartik Mishra
- Subjects
Protozoan Vaccines ,Science ,Trypanosoma cruzi ,Protein subunit ,Plasmodium falciparum ,Epitopes, T-Lymphocyte ,Virulence ,Computational biology ,Article ,Epitope ,Cholera ,Antigen ,MHC class I ,Humans ,Chagas Disease ,Malaria, Falciparum ,Vibrio cholerae ,Vaccines ,Multidisciplinary ,biology ,Immunogenicity ,Reverse vaccinology ,Computational Biology ,Computational biology and bioinformatics ,Vaccinology ,Bacterial vaccine ,Bacterial Vaccines ,biology.protein ,Medicine ,Epitopes, B-Lymphocyte - Abstract
Antigen identification is an important step in the vaccine development process. Computational approaches including deep learning systems can play an important role in the identification of vaccine targets using genomic and proteomic information. Here, we present a new computational system to discover and analyse novel vaccine targets leading to the design of a multi-epitope subunit vaccine candidate. The system incorporates reverse vaccinology and immuno-informatics tools to screen genomic and proteomic datasets of several pathogens such as Trypanosoma cruzi, Plasmodium falciparum, and Vibrio cholerae to identify potential vaccine candidates (PVC). Further, as a case study, we performed a detailed analysis of the genomic and proteomic dataset of T. cruzi (CL Brenner and Y strain) to shortlist eight proteins as possible vaccine antigen candidates using properties such as secretory/surface-exposed nature, low transmembrane helix (Trypanosoma species and strains. Further, studies are required to validate safety and immunogenicity of the vaccine.
- Published
- 2021
- Full Text
- View/download PDF
17. Signal Transducer and Activator of Transcription-3 Modulation of Cardiac Pathology in Chronic Chagasic Cardiomyopathy
- Author
-
Kristyn A. Hoffman, Maria Jose Villar, Cristina Poveda, Maria Elena Bottazzi, Peter J. Hotez, David J. Tweardy, and Kathryn M. Jones
- Subjects
Chagas Cardiomyopathy ,STAT3 Transcription Factor ,Microbiology (medical) ,Cardiac function curve ,medicine.medical_specialty ,Cardiac fibrosis ,Trypanosoma cruzi ,Immunology ,Microbiology ,STAT3 ,Mice ,Cellular and Infection Microbiology ,Fibrosis ,Internal medicine ,medicine ,Animals ,Chagas Disease ,STAT1 ,Original Research ,biology ,business.industry ,fibrosis ,Heart ,Dilated cardiomyopathy ,medicine.disease ,chronic Chagasic cardiomyopathy ,QR1-502 ,Chronic infection ,Infectious Diseases ,inflammation ,Benznidazole ,Heart failure ,biology.protein ,Cardiology ,business ,medicine.drug - Abstract
Chronic Chagasic cardiomyopathy (CCC) is a severe clinical manifestation that develops in 30%–40% of individuals chronically infected with the protozoal parasite Trypanosoma cruzi and is thus an important public health problem. Parasite persistence during chronic infection drives pathologic changes in the heart, including myocardial inflammation and progressive fibrosis, that contribute to clinical disease. Clinical manifestations of CCC span a range of symptoms, including cardiac arrhythmias, thromboembolic disease, dilated cardiomyopathy, and heart failure. This study aimed to investigate the role of signal transducer and activator of transcription-3 (STAT3) in cardiac pathology in a mouse model of CCC. STAT3 is a known cellular mediator of collagen deposition and fibrosis. Mice were infected with T. cruzi and then treated daily from 70 to 91 days post infection (DPI) with TTI-101, a small molecule inhibitor of STAT3; benznidazole; a combination of benznidazole and TTI-101; or vehicle alone. Cardiac function was evaluated at the beginning and end of treatment by echocardiography. By the end of treatment, STAT3 inhibition with TTI-101 eliminated cardiac fibrosis and fibrosis biomarkers but increased cardiac inflammation; serum levels of interleukin-6 (IL-6), and IFN−γ; cardiac gene expression of STAT1 and nuclear factor-κB (NF-κB); and upregulation of IL-6 and Type I and Type II IFN responses. Concurrently, decreased heart function was measured by echocardiography and myocardial strain. These results indicate that STAT3 plays a critical role in the cardiac inflammatory–fibrotic axis during CCC.
- Published
- 2021
- Full Text
- View/download PDF
18. Differential expression and regulation of MS4A family members in myeloid cells in physiological and pathological conditions
- Author
-
Fabio Grizzi, Alberto Mantovani, Domenico Supino, Fabio Pasqualini, Marie-Astrid Boutet, Andrea Gianatti, Marina Sironi, Maria José Oliveira, Barbara Bottazzi, Silvia Carnevale, Matteo Stravalaci, Sarah N. Mapelli, Irene Mattiola, C. Pitzalis, Rémi Porte, Rita Silva-Gomes, Federico Colombo, Massimo Locati, Humanitas University [Milan] (Hunimed), Universidade do Porto, Istituto Clinico Humanitas [Milan] (IRCCS Milan), Regenerative Medicine and Skeleton research lab (RMeS), Ecole Nationale Vétérinaire, Agroalimentaire et de l'alimentation Nantes-Atlantique (ONIRIS)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM), Queen Mary University of London (QMUL), Berlin Institute of Health (BIH), Charité - UniversitätsMedizin = Charité - University Hospital [Berlin], Mucosal and Developmental Immunology [Berlin, Germany], IRCCS San Raffaele Scientific Institute [Milan, Italie], Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Azienda Ospedaliera Ospedale Papa Giovanni XXIII [Bergamo, Italy], University of Milan, Federal University of Health Sciences of Porto Alegre = Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Universidade do Porto = University of Porto, Regenerative Medicine and Skeleton (RMeS), École nationale vétérinaire, agroalimentaire et de l'alimentation Nantes-Atlantique (ONIRIS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN), Università degli Studi di Milano = University of Milan (UNIMI), and Jehan, Frederic
- Subjects
rheumatoid arthritis ,[SDV.MHEP.AHA] Life Sciences [q-bio]/Human health and pathology/Tissues and Organs [q-bio.TO] ,Myeloid ,Immunology ,Population ,Biology ,Monocytes ,03 medical and health sciences ,0302 clinical medicine ,monocytes/Mϕs ,medicine ,[SDV.MHEP.AHA]Life Sciences [q-bio]/Human health and pathology/Tissues and Organs [q-bio.TO] ,Immunology and Allergy ,Gene family ,Humans ,Family ,education ,030304 developmental biology ,Regulation of gene expression ,[SDV.MHEP.RSOA] Life Sciences [q-bio]/Human health and pathology/Rhumatology and musculoskeletal system ,0303 health sciences ,education.field_of_study ,MS4A6A ,[SDV.MHEP.GEG] Life Sciences [q-bio]/Human health and pathology/Geriatry and gerontology ,[SDV.MHEP.GEG]Life Sciences [q-bio]/Human health and pathology/Geriatry and gerontology ,MS4A3 ,COVID-19 ,Membrane Proteins ,Cell Biology ,Antigens, CD20 ,In vitro ,3. Good health ,Cell biology ,MS4A4A ,medicine.anatomical_structure ,[SDV.MHEP.RSOA]Life Sciences [q-bio]/Human health and pathology/Rhumatology and musculoskeletal system ,MS4A2 ,030217 neurology & neurosurgery ,Function (biology) - Abstract
The MS4A gene family encodes 18 tetraspanin-like proteins, most of which with unknown function. MS4A1 (CD20), MS4A2 (FcεRIβ), MS4A3 (HTm4), and MS4A4A play important roles in immunity, whereas expression and function of other members of the family are unknown. The present investigation was designed to obtain an expression fingerprint of MS4A family members, using bioinformatics analysis of public databases, RT-PCR, and protein analysis when possible. MS4A3, MS4A4A, MS4A4E, MS4A6A, MS4A7, and MS4A14 were expressed by myeloid cells. MS4A6A and MS4A14 were expressed in circulating monocytes and decreased during monocyte-to-Mϕ differentiation in parallel with an increase in MS4A4A expression. Analysis of gene expression regulation revealed a strong induction of MS4A4A, MS4A6A, MS4A7, and MS4A4E by glucocorticoid hormones. Consistently with in vitro findings, MS4A4A and MS4A7 were expressed in tissue Mϕs from COVID-19 and rheumatoid arthritis patients. Interestingly, MS4A3, selectively expressed in myeloid precursors, was found to be a marker of immature circulating neutrophils, a cellular population associated to COVID-19 severe disease. The results reported here show that members of the MS4A family are differentially expressed and regulated during myelomonocytic differentiation, and call for assessment of their functional role and value as therapeutic targets.
- Published
- 2021
- Full Text
- View/download PDF
19. The potential role of Th17 immune responses in coronavirus immunopathology and vaccine-induced immune enhancement
- Author
-
David B. Corry, Maria Elena Bottazzi, and Peter J. Hotez
- Subjects
0301 basic medicine ,COVID-19 Vaccines ,medicine.medical_treatment ,Pneumonia, Viral ,030106 microbiology ,Immunology ,Biology ,medicine.disease_cause ,Microbiology ,Betacoronavirus ,03 medical and health sciences ,Immune system ,Immunopathology ,Eosinophilic ,medicine ,Humans ,Pandemics ,Coronavirus ,Respiratory tract infections ,Interleukin-6 ,SARS-CoV-2 ,COVID-19 ,Viral Vaccines ,medicine.disease ,Vaccination ,Pneumonia ,030104 developmental biology ,Infectious Diseases ,Cytokine ,Gene Expression Regulation ,Cytokines ,Th17 Cells ,Coronavirus Infections - Abstract
Increasing evidence points to host Th17 inflammatory responses as contributing to the severe lung pathology and mortality of lower respiratory tract infections from coronaviruses. This includes host inflammatory and cytokine responses to COVID-19 caused by the SARS-2 coronavirus (SARS CoV2). From studies conducted in laboratory animals, there are additional concerns about immune enhancement and the role of potential host immunopathology resulting from experimental human COVID-19 vaccines. Here we summarize evidence suggesting there may be partial overlap between the underlying immunopathologic processes linked to both coronavirus infection and vaccination, and a role for Th17 in immune enhancement and eosinophilic pulmonary immunopathology. Such findings help explain the link between viral-vectored coronavirus vaccines and immune enhancement and its reduction through alum adjuvants. Additional research may also clarify links between COVID-19 pulmonary immunopathology and heart disease.
- Published
- 2020
- Full Text
- View/download PDF
20. A phase 1 study of the safety, reactogenicity, and immunogenicity of a Schistosoma mansoni vaccine with or without glucopyranosyl lipid A aqueous formulation (GLA-AF) in healthy adults from a non-endemic area
- Author
-
G.E. Potter, W. Jones, Maria Elena Bottazzi, H. M. El Sahly, Shital M. Patel, Jordan L. Plieskatt, Jeffrey M. Bethony, Wendy A. Keitel, Peter J. Hotez, J.K. Kennedy, Gregory A. Deye, Robert L. Atmar, and David Diemert
- Subjects
Male ,medicine.medical_treatment ,Gastroenterology ,Cohort Studies ,Immunogenicity, Vaccine ,0302 clinical medicine ,Glucosides ,Schistosomiasis ,Medicine ,030212 general & internal medicine ,Vaccines ,education.field_of_study ,biology ,Immunogenicity ,Schistosoma mansoni ,Middle Aged ,Healthy Volunteers ,Lipid A ,Infectious Diseases ,Cytokines ,Molecular Medicine ,Female ,Adjuvant ,Adult ,medicine.medical_specialty ,Adolescent ,030231 tropical medicine ,Population ,Antibodies, Helminth ,Placebo ,Article ,Young Adult ,03 medical and health sciences ,Adjuvants, Immunologic ,Double-Blind Method ,Internal medicine ,Animals ,Humans ,education ,Reactogenicity ,Dose-Response Relationship, Drug ,General Veterinary ,General Immunology and Microbiology ,business.industry ,Public Health, Environmental and Occupational Health ,Tropical disease ,medicine.disease ,biology.organism_classification ,Antigens, Helminth ,Immunoglobulin G ,business - Abstract
BACKGROUND: Schistosomiasis caused by Schistosoma mansoni (Sm) is a chronic, debilitating and potentially deadly neglected tropical disease. The licensure of a vaccine to prevent schistosomiasis would represent a major breakthrough in public health. METHODS: The safety and immunogenicity of a candidate Sm vaccine were assessed in this phase I, double-blind, dose-escalation trial. Seventy-two healthy Sm-naïve 18–50 year olds were randomized to receive 3 doses~ 8 weeks apart of saline placebo, or 10 μg, 30 μg, or 100 μg of recombinant Sm-Tetraspanin-2 vaccine formulated on aluminum hydroxide adjuvant (Sm-TSP-2/Al) with or without 5 μg of glucopyranosyl lipid A aqueous formulation (GLA-AF). Clinical and serologic responses were assessed for 1 year after dose 3. RESULTS: Vaccines were safe and well-tolerated. The most common reactions were injection site tenderness and pain, and headache and fatigue. Tenderness and pain were more frequent in groups receiving vaccine with GLA-AF than placebo (p = 0.0036 and p = 0.0014, respectively). Injection site reactions among those given Sm-TSP-2/Al with GLA-AF lasted 1.22 and 1.33 days longer than those receiving Sm-TSP-2/Al without GLA-AF or placebo (p < 0.001 for both). Dose- and adjuvant-related increases in serum IgG against Sm-TSP-2 were observed. Peak IgG levels occurred 14 days after dose 3. Seroresponse frequencies were low among recipients of Sm-TSP-2/Al without GLA-AF, but higher among subjects receiving 30 μg or 100 μg of Sm-TSP-2/Al with GLA-AF. More seroresponses were observed among those given 30 μg or 100 μg of Sm-TSP-2/Al with GLA-AF compared to placebo (p = 0.023 and p < 0.001, respectively). Seroresponse frequencies were 0%, 30%, 50%, and 89%, respectively, among those given placebo, or 10 μg, 30 μg or 100 μg of Sm-TSP-2/Al with GLA-AF, suggesting a dose-response relationship for Sm-TSP-2/Al with GLA-AF (p = 0.0001). CONCLUSIONS: Sm-TSP-2/Al with or without GLA-AF was safe and well tolerated in a Sm-naïve population. A vaccine like the one under development may represent our best hope to eliminating this neglected tropical disease.
- Published
- 2019
- Full Text
- View/download PDF
21. Receptor-binding domain recombinant protein on alum-CpG induces broad protection against SARS-CoV-2 variants of concern
- Author
-
Rakhi Tyagi Kundu, Ulrich Strych, Peter J. Hotez, Wen-Hsiang Chen, Shannon E. Ronca, Vikram Paradkar, Maria Jose Villar, Jeroen Pollet, Martin Reers, Cristina Poveda, Rakesh Adhikari, Brian Keegan, Leroy Versteeg, Jason T. Kimata, Jungsoon Lee, Portia M. Gillespie, Bin Zhan, Junfei Wei, Zhuyun Liu, Maria Elena Bottazzi, and Brianna Lopez
- Subjects
COVID-19 Vaccines ,chemical and pharmacologic phenomena ,Booster dose ,Antibodies, Viral ,complex mixtures ,Article ,law.invention ,chemistry.chemical_compound ,Mice ,Immune system ,law ,Animals ,Humans ,Neutralizing antibody ,COVID-19 Serotherapy ,biology ,General Veterinary ,General Immunology and Microbiology ,Chemistry ,Alum ,SARS-CoV-2 ,Public Health, Environmental and Occupational Health ,Immunization, Passive ,COVID-19 ,Virology ,Antibodies, Neutralizing ,Recombinant Proteins ,Titer ,Infectious Diseases ,CpG site ,Spike Glycoprotein, Coronavirus ,biology.protein ,Recombinant DNA ,Molecular Medicine ,Alum Compounds ,Antibody - Abstract
We conducted preclinical studies in mice using a yeast-produced SARS-CoV-2 RBD219-N1C1 subunit vaccine candidate formulated with aluminum hydroxide (alum) and CpG deoxynucleotides. This vaccine formulation is similar to one that entered advanced phase 3 clinical development in India. We compared the immune response of mice vaccinated with RBD219-N1C1/alum to mice vaccinated with RBD219-N1C1/alum+CpG. We also evaluated mice immunized with RBD219-N1C1/alum+CpG and boosted with RBD219-N1C1/alum. Mice were immunized twice intramuscularly at a 21-day interval. Compared to two doses of the RBD219-N1C1/alum formulation, the RBD219-N1C1/alum+CpG vaccine induced a stronger and more balanced Th1/Th2 cellular immune response, with high levels of neutralizing antibodies against the original Wuhan isolate of SARS-CoV-2 as well as the B.1.1.7 (Alpha), B.1.351 (Beta) and B.1.617.1 (Kappa) variants. Notably, the sera from mice that received two 7 µg doses of RBD219-N1C1/alum+CpG showed more than 18 times higher neutralizing antibody titers against B.1.351, than the WHO International Standard for anti-SARS-CoV-2 immunoglobulin NIBSC 20/136. Interestingly, a booster dose did not require the addition of CpG to induce this effect. The data reported here reinforces that the RBD219-N1C1/alum+CpG vaccine formulation is suitable for inducing broadly neutralizing antibodies against SARS-CoV-2 including three variants of concern, B.1.1.7 (Alpha), B.1.351 (Beta), and B.1.617.1 (Kappa).
- Published
- 2021
22. Recognition and inhibition of SARS-CoV-2 by humoral innate immunity pattern recognition molecules
- Author
-
Milos Matkovic, Francesco Scavello, Mattia Pedotti, Andrea Cavalli, Elisa Vicenzi, Daniela Cesana, Sarah N. Mapelli, Nicasio Mancini, Valeria Capurro, Stefano Duga, Alberto Mantovani, Rino Rappuoli, Rosanna Asselta, Marina Sironi, Pietro Invernizzi, Cecilia Garlanda, Mariagrazia Uguccioni, Luca Varani, Pierangela Gallina, Elvezia Maria Paraboschi, Nicola Clementi, Matteo Stravalaci, Isabel Pagani, Andrea Doni, Nicoletta Pedemonte, and Barbara Bottazzi
- Subjects
Glycan ,Innate immune system ,biology ,business.industry ,viruses ,Pattern recognition ,PTX3 ,In vitro ,Nucleoprotein ,Complement system ,Lectin pathway ,biology.protein ,Artificial intelligence ,business ,Mannan-binding lectin - Abstract
SummaryThe humoral arm of innate immunity includes diverse molecules with antibody-like functions, some of which serve as disease severity biomarkers in COVID-19. The present study was designed to conduct a systematic investigation of the interaction of humoral fluid phase pattern recognition molecules (PRM) with SARS-CoV-2. Out of 10 PRM tested, the long pentraxin PTX3 and Mannose Binding Lectin (MBL) bound the viral Nucleoprotein and Spike, respectively. MBL bound trimeric Spike, including that of variants of concern, in a glycan- dependent way and inhibited SARS-CoV-2 in threein vitromodels. Moreover, upon binding to Spike, MBL activated the lectin pathway of complement activation. Genetic polymorphisms at the MBL locus were associated with disease severity. These results suggest that selected humoral fluid phase PRM can play an important role in resistance to, and pathogenesis of, COVID-19, a finding with translational implications.
- Published
- 2021
- Full Text
- View/download PDF
23. Alum:CpG adjuvant enables SARS-CoV-2 RBD-induced protection in aged mice and synergistic activation of human elder type 1 immunity
- Author
-
Etsuro Nanishi, Francesco Borriello, Timothy R. O’Meara, Marisa E. McGrath, Yoshine Saito, Robert E. Haupt, Hyuk-Soo Seo, Simon D. van Haren, Byron Brook, Jing Chen, Joann Diray-Arce, Simon Doss-Gollin, Maria De Leon, Katherine Chew, Manisha Menon, Kijun Song, Andrew Z. Xu, Timothy M. Caradonna, Jared Feldman, Blake M. Hauser, Aaron G. Schmidt, Amy C. Sherman, Lindsey R. Baden, Robert K. Ernst, Carly Dillen, Stuart M. Weston, Robert M. Johnson, Holly L. Hammond, Romana Mayer, Allen Burke, Maria E. Bottazzi, Peter J. Hotez, Ulrich Strych, Aiquan Chang, Jingyou Yu, Dan H. Barouch, Sirano Dhe-Paganon, Ivan Zanoni, Al Ozonoff, Matthew B. Frieman, Ofer Levy, and David J. Dowling
- Subjects
Chemokine ,COVID-19 Vaccines ,CpG Oligodeoxynucleotide ,Aluminum Hydroxide ,Context (language use) ,Antibodies, Viral ,Article ,Mice ,Animals ,Humans ,Medicine ,Pandemics ,BNT162 Vaccine ,Aged ,Vaccines, Synthetic ,biology ,SARS-CoV-2 ,business.industry ,Immunogenicity ,COVID-19 ,TLR9 ,Antibodies, Neutralizing ,CpG site ,Spike Glycoprotein, Coronavirus ,Immunology ,TLR3 ,biology.protein ,mRNA Vaccines ,Antibody ,business - Abstract
Global deployment of vaccines that can provide protection across several age groups is still urgently needed to end the COVID-19 pandemic especially for low- and middle-income countries. While vaccines against SARS-CoV-2 based on mRNA and adenoviral-vector technologies have been rapidly developed, additional practical and scalable SARS-CoV-2 vaccines are needed to meet global demand. In this context, protein subunit vaccines formulated with appropriate adjuvants represent a promising approach to address this urgent need. Receptor-binding domain (RBD) is a key target of neutralizing antibodies (Abs) but is poorly immunogenic. We therefore compared pattern recognition receptor (PRR) agonists, including those activating STING, TLR3, TLR4 and TLR9, alone or formulated with aluminum hydroxide (AH), and benchmarked them to AS01B and AS03-like emulsion-based adjuvants for their potential to enhance RBD immunogenicity in young and aged mice. We found that the AH and CpG adjuvant formulation (AH:CpG) demonstrated the highest enhancement of anti-RBD neutralizing Ab titers in both age groups (∼80-fold over AH), and protected aged mice from the SARS-CoV-2 challenge. Notably, AH:CpG-adjuvanted RBD vaccine elicited neutralizing Abs against both wild-type SARS-CoV-2 and B.1.351 variant at serum concentrations comparable to those induced by the authorized mRNA BNT162b2 vaccine. AH:CpG induced similar cytokine and chemokine gene enrichment patterns in the draining lymph nodes of both young adult and aged mice and synergistically enhanced cytokine and chemokine production in human young adult and elderly mononuclear cells. These data support further development of AH:CpG-adjuvanted RBD as an affordable vaccine that may be effective across multiple age groups.One Sentence SummaryAlum and CpG enhance SARS-CoV-2 RBD protective immunity, variant neutralization in aged mice and Th1-polarizing cytokine production by human elder leukocytes.
- Published
- 2021
- Full Text
- View/download PDF
24. Severity of SARS-CoV-2 infection as a function of the interferon landscape across the respiratory tract of COVID-19 patients
- Author
-
Andrea Bottazzi, Alessandro Ambrosi, Roberto Ferrarese, Sofia Sisti, Antonio E. Pontiroli, Laura Saracino, Fabio A. Facchini, Benedetta Sposito, Laura Pandolfi, Andreas Wack, Laura Marongiu, Vanessa Frangipane, Massimo Clementi, Ivan Zanoni, Stefania Crotta, Enju Liu, Nicasio Mancini, Riccardo Colombo, Federica Meloni, Elena Tagliabue, Tommaso Fossali, Nicola Clementi, and Achille Broggi
- Subjects
Cell type ,medicine.anatomical_structure ,Coronavirus disease 2019 (COVID-19) ,Interferon ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Immunology ,medicine ,Outbreak ,Biology ,Viral load ,Function (biology) ,Respiratory tract ,medicine.drug - Abstract
SummaryThe COVID-19 outbreak driven by SARS-CoV-2 has caused more than 2.5 million deaths globally, with the most severe cases characterized by over-exuberant production of immune-mediators, the nature of which is not fully understood. Interferons of the type I (IFN-I) or type III (IFN-III) families are potent antivirals, but their role in COVID-19 remains debated. Our analysis of gene and protein expression along the respiratory tract shows that IFNs, especially IFN-III, are over-represented in the lower airways of patients with severe COVID-19, while high levels of IFN-III, and to a lesser extent IFN-I, characterize the upper airways of patients with high viral burden but reduced disease risk or severity; also, IFN expression varies with abundance of the cell types that produce them. Our data point to a dynamic process of inter- and intra-family production of IFNs in COVID-19, and suggest that IFNs play opposing roles at distinct anatomical sites.
- Published
- 2021
- Full Text
- View/download PDF
25. PTX3 Regulation of Inflammation, Hemostatic Response, Tissue Repair, and Resolution of Fibrosis Favors a Role in Limiting Idiopathic Pulmonary Fibrosis
- Author
-
Andrea Doni, Alberto Mantovani, Barbara Bottazzi, and Remo Castro Russo
- Subjects
0301 basic medicine ,Extracellular matrix ,Idiopathic pulmonary fibrosis ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Fibrosis ,humoral immunity ,Immunology and Allergy ,biology ,Interstitial lung disease ,respiratory system ,Extracellular Matrix ,Serum Amyloid P-Component ,C-Reactive Protein ,030220 oncology & carcinogenesis ,Perspective ,medicine.symptom ,Immunology ,Inflammation ,Nerve Tissue Proteins ,Bleomycin ,Fibrin ,03 medical and health sciences ,medicine ,Animals ,Humans ,PTX3 ,Hemostasis ,Wound Healing ,Innate immune system ,business.industry ,fibrosis ,resolution ,Plasminogen ,RC581-607 ,medicine.disease ,Idiopathic Pulmonary Fibrosis ,Immunity, Innate ,respiratory tract diseases ,Immunity, Humoral ,Disease Models, Animal ,030104 developmental biology ,IPF ,chemistry ,Cancer research ,biology.protein ,Immunologic diseases. Allergy ,business - Abstract
PTX3 is a soluble pattern recognition molecule (PRM) belonging to the humoral innate immune system, rapidly produced at inflammatory sites by phagocytes and stromal cells in response to infection or tissue injury. PTX3 interacts with microbial moieties and selected pathogens, with molecules of the complement and hemostatic systems, and with extracellular matrix (ECM) components. In wound sites, PTX3 interacts with fibrin and plasminogen and favors a timely removal of fibrin-rich ECM for an efficient tissue repair. Idiopathic Pulmonary Fibrosis (IPF) is a chronic and progressive interstitial lung disease of unknown origin, associated with excessive ECM deposition affecting tissue architecture, with irreversible loss of lung function and impact on the patient’s life quality. Maccarinelli et al. recently demonstrated a protective role of PTX3 using the bleomycin (BLM)-induced experimental model of lung fibrosis, in line with the reported role of PTX3 in tissue repair. However, the mechanisms and therapeutic potential of PTX3 in IPF remained to be investigated. Herein, we provide new insights on the possible role of PTX3 in the development of IPF and BLM-induced lung fibrosis. In mice, PTX3-deficiency was associated with worsening of the disease and with impaired fibrin removal and subsequently increased collagen deposition. In IPF patients, microarray data indicated a down-regulation of PTX3 expression, thus suggesting a potential rational underlying the development of disease. Therefore, we provide new insights for considering PTX3 as a possible target molecule underlying therapeutic intervention in IPF.
- Published
- 2021
26. A yeast expressed RBD-based SARS-CoV-2 vaccine formulated with 3M-052-alum adjuvant promotes protective efficacy in non-human primates
- Author
-
Guido Ferrari, Neeta Shenvi, Thomas H. Vanderford, Susan Pereira Ribeiro, Wen-Hsiang Chen, David C. Montefiori, Talha Abid, Alessandro Sette, Daniela Weiskopf, Debashis Dutta, Katharine Floyd, Shelly Wang, Dieter Mielke, Sudhir Pai Kasturi, Georgia D. Tomaras, Pamela A. Kozlowski, Celia C. LaBranche, Maria Pino, Jungsoon Lee, Sherrie Jean, Venkata Viswanadh Edara, Xiaoying Shen, Justin Pollara, Justin C Smith, Mirko Paiardini, Rafick Pierre Sekaly, Fawn Connor-Stroud, Joyce Cohen, Gabriela Pacheco-Sanchez, Hongmei Gao, Zhuyun Liu, Christopher B. Fox, Sanjeev Gumber, Junfei Wei, Nathan Eisel, Maria Elena Bottazzi, Rachelle L. Stammen, Jennifer S. Wood, Shannon Kirejczyk, Bin Zhan, Muhammad Bilal Latif, Peter J. Hotez, Kirk Easley, Ulrich Strych, Jeroen Pollet, Mehul S. Suthar, and Mark A. Tomai
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,Male ,Immunogen ,COVID-19 Vaccines ,T cell ,Immunology ,Biology ,CD8-Positive T-Lymphocytes ,Antibodies, Viral ,Immunoglobulin G ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Adjuvants, Immunologic ,Protein Domains ,Administration, Inhalation ,medicine ,Animals ,Humans ,Alum adjuvant ,Lung ,Administration, Intranasal ,SARS-CoV-2 ,COVID-19 ,General Medicine ,Viral Load ,Antibodies, Neutralizing ,Macaca mulatta ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Saccharomycetales ,Spike Glycoprotein, Coronavirus ,biology.protein ,Alum Compounds ,Cytokines ,Nasal administration ,Antibody ,Viral load ,Protein Binding - Abstract
Ongoing SARS-CoV-2 vaccine development is focused on identifying stable, cost-effective, and accessible candidates for global use, specifically in low and middle-income countries. Here, we report the efficacy of a rapidly scalable, novel yeast expressed SARS-CoV-2 specific receptor-binding domain (RBD) based vaccine in rhesus macaques. We formulated the RBD immunogen in alum, a licensed and an emerging alum adsorbed TLR-7/8 targeted, 3M-052-alum adjuvants. The RBD+3M-052-alum adjuvanted vaccine promoted better RBD binding and effector antibodies, higher CoV-2 neutralizing antibodies, improved Th1 biased CD4+T cell reactions, and increased CD8+ T cell responses when compared to the alum-alone adjuvanted vaccine. RBD+3M-052-alum induced a significant reduction of SARS-CoV-2 virus in respiratory tract upon challenge, accompanied by reduced lung inflammation when compared with unvaccinated controls. Anti-RBD antibody responses in vaccinated animals inversely correlated with viral load in nasal secretions and BAL. RBD+3M-052-alum blocked a post SARS-CoV-2 challenge increase in CD14+CD16++ intermediate blood monocytes, and Fractalkine, MCP-1, and TRAIL in the plasma. Decreased plasma analytes and intermediate monocyte frequencies correlated with reduced nasal and BAL viral loads. Lastly, RBD-specific plasma cells accumulated in the draining lymph nodes and not in the bone marrow, contrary to previous findings. Together, these data show that a yeast expressed, RBD-based vaccine+3M-052-alum provides robust immune responses and protection against SARS-CoV-2, making it a strong and scalable vaccine candidate.
- Published
- 2021
27. Circulating biomarkers and cardiac function over 3 years after chemotherapy with anthracyclines: the ICOS-ONE trial
- Author
-
Barbara Bottazzi, Giuseppe Curigliano, Maria Teresa Sandri, Roberto Leone, Jennifer Meessen, Pietro Cortesi, Daniele Nassiacos, Enrico Nicolis, Alessandra Bianchi, Deborah Novelli, Alberto Farolfi, Icos‐One Study Investigators, Cecilia Garlanda, Giovanna Balconi, Alessandro Colombo, Marco Bregni, Enrico Barbieri, Roberto Latini, Daniela Cardinale, Carlo M. Cipolla, Maurizio Civelli, Paolo Pastori, Stefania Gori, Lidia Staszewsky, Alberto Mantovani, Serge Masson, Carlo Milandri, Fabio Ciceri, Francesco Ghisoni, Gianluigi Condorelli, Michela Salvatici, Claudio Verusio, Alessandra Malossi, Maria Grazia Franzosi, Cristina Falci, GianFranco Cucchi, Maurizio Mangiavacchi, Anna Monopoli, Elisabetta Menatti, Meessen, J. M. T. A., Cardinale, D., Ciceri, F., Sandri, M. T., Civelli, M., Bottazzi, B., Cucchi, G., Menatti, E., Mangiavacchi, M., Condorelli, G., Barbieri, E., Gori, S., Colombo, A., Curigliano, G., Salvatici, M., Pastori, P., Ghisoni, F., Bianchi, A., Falci, C., Cortesi, P., Farolfi, A., Monopoli, A., Milandri, C., Bregni, M., Malossi, A., Nassiacos, D., Verusio, C., Staszewsky, L., Leone, R., Novelli, D., Balconi, G., Nicolis, E. B., Franzosi, M. G., Masson, S., Garlanda, C., Mantovani, A., Cipolla, C. M., and Latini, R.
- Subjects
Cardiac function curve ,lcsh:Diseases of the circulatory (Cardiovascular) system ,medicine.medical_specialty ,030204 cardiovascular system & hematology ,Inducible T-Cell Co-Stimulator Protein ,03 medical and health sciences ,Ventricular Dysfunction, Left ,0302 clinical medicine ,Heart arrhythmia ,Internal medicine ,Original Research Articles ,Troponin I ,Natriuretic Peptide, Brain ,medicine ,Humans ,Anthracyclines ,030212 general & internal medicine ,Original Research Article ,Ejection fraction ,biology ,business.industry ,medicine.disease ,Troponin ,Pulmonary embolism ,Cardio‐oncology ,Cardio-oncology ,lcsh:RC666-701 ,Echocardiography ,Heart failure ,Cardiology ,biology.protein ,Cardiac dysfunction ,Cardiology and Cardiovascular Medicine ,business ,Biomarkers ,Blood sampling - Abstract
Aims: A multicentre trial, ICOS-ONE, showed increases above the upper limit of normality of cardiac troponin (cTn) in 27% of patients within 12months after the end of cancer chemotherapy (CT) with anthracyclines, whether cardiac protection with enalapril was started at study entry in all (prevention arm) or only upon first occurrence on supra-normal cTn (troponin-triggered arm). The aims of the present post hoc analysis were (i) to assess whether anthracycline-based treatment could induce cardiotoxicity over 36month follow-up and (ii) to describe the time course of three cardiovascular biomarkers (i.e. troponin I cTnI-Ultra, B-type natriuretic peptide BNP, and pentraxin 3 PTX3) and of left ventricular (LV) function up to 36months. Methods and results: Eligible patients were those prescribed first-in-life CT, without evidence of cardiovascular disease, normal cTn, LV ejection fraction (EF) >50%, not on renin-angiotensin aldosterone system antagonists. Patients underwent echocardiography and blood sampling at 24 and 36months. No differences were observed in biomarker concentration between the two study arms, ‘prevention' vs. ‘troponin-triggered'. During additional follow-up 13 more deaths occurred, leading to a total of 23 (9.5%), all due to a non-cardiovascular cause. No new occurrences of LV-dysfunction were reported. Two additional patients were admitted to the hospital for cardiovascular causes, both for acute pulmonary embolism. No first onset of raised cTnI-Ultra was reported in the extended follow-up. BNP remained within normal range: at 36months was 23.4ng/L, higher (N.S.) than at baseline, 17.6ng/L. PTX3 peaked at 5.2ng/mL 1month after CT and returned to baseline values thereafter. cTnI-Ultra peaked at 26ng/L 1month after CT and returned to 3ng/L until the last measurement at 36months. All echocardiographic variables remained stable during follow-up with a median LVEF of 63% and left atrial volume index about 24mL/m2. Conclusions: First-in-life CT with median cumulative dose of anthracyclines of 180mg/m2 does not seem to cause clinically significant cardiac injury, as assessed by circulating biomarkers and echocardiography, in patients aged 51years (median), without pre-existing cardiac disease. This may suggest either a 100% efficacy of enalapril (given as preventive or troponin-triggered) or a reassuringly low absolute cardiovascular risk in this cohort of patients, which may not require intensive cardiologic follow-up.
- Published
- 2020
28. Process development and scale-up optimization of the SARS-CoV-2 receptor binding domain-based vaccine candidate, RBD219-N1C1
- Author
-
Jungsoon Lee, Rakhi Tyagi Kundu, Joanne Altieri Rivera, Bin Zhan, Wen-Hsiang Chen, Peter J. Hotez, Zhuyun Liu, Junfei Wei, Maria Elena Bottazzi, Rakesh Adhikari, Portia M. Gillespie, and Ulrich Strych
- Subjects
COVID-19 Vaccines ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Spike protein ,Applied Microbiology and Biotechnology ,Pichia pastoris ,law.invention ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antigen ,law ,Animals ,Humans ,030212 general & internal medicine ,Neutralizing antibody ,Purification ,030304 developmental biology ,0303 health sciences ,biology ,Chemistry ,SARS-CoV-2 ,COVID-19 ,Reproducibility of Results ,General Medicine ,biology.organism_classification ,Yeast ,Biotechnological Products and Process Engineering ,Biochemistry ,Yield (chemistry) ,SCALE-UP ,Saccharomycetales ,Spike Glycoprotein, Coronavirus ,Fermentation ,biology.protein ,Recombinant DNA ,Biotechnology - Abstract
Abstract A SARS-CoV-2 RBD219-N1C1 (RBD219-N1C1) recombinant protein antigen formulated on Alhydrogel® has recently been shown to elicit a robust neutralizing antibody response against SARS-CoV-2 pseudovirus in mice. The antigen has been produced under current good manufacturing practices (cGMPs) and is now in clinical testing. Here, we report on process development and scale-up optimization for upstream fermentation and downstream purification of the antigen. This includes production at the 1-L and 5-L scales in the yeast, Pichia pastoris, and the comparison of three different chromatographic purification methods. This culminated in the selection of a process to produce RBD219-N1C1 with a yield of >400 mg per liter of fermentation with >92% purity and >39% target product recovery after purification. In addition, we show the results from analytical studies, including SEC-HPLC, DLS, and an ACE2 receptor binding assay that were performed to characterize the purified proteins to select the best purification process. Finally, we propose an optimized upstream fermentation and downstream purification process that generates quality RBD219-N1C1 protein antigen and is fully scalable at a low cost. Key points • Yeast fermentation conditions for a recombinant COVID-19 vaccine were determined. • Three purification protocols for a COVID-19 vaccine antigen were compared. • Reproducibility of a scalable, low-cost process for a COVID-19 vaccine was shown. Graphical abstract
- Published
- 2021
29. Serum amyloid P component is an essential element of resistance against Aspergillus fumigatus
- Author
-
Agostinho Carvalho, Johan Maertens, Marina Botto, Toine Mercier, Tilo Schorn, João F. Lacerda, Barbara Bottazzi, Katrien Lagrou, Ilaria Laface, António Campos, Sarah N. Mapelli, Francesca Petroni, Elena Magrini, Alberto Mantovani, Antonio Inforzato, John D. Lambris, Cristina Cunha, Raffaella Parente, Federico Colombo, Rémi Porte, Andrea Doni, Cecilia Garlanda, and Repositório da Universidade de Lisboa
- Subjects
0301 basic medicine ,Fungal infection ,genetic structures ,Neutrophils ,Science ,Phagocytosis ,General Physics and Astronomy ,ComputingMilieux_LEGALASPECTSOFCOMPUTING ,General Biochemistry, Genetics and Molecular Biology ,Article ,Aspergillus fumigatus ,Microbiology ,03 medical and health sciences ,Classical complement pathway ,Immunocompromised Host ,Mice ,0302 clinical medicine ,mental disorders ,Animals ,Humans ,skin and connective tissue diseases ,Opsonin ,Lung ,Serum amyloid P component ,Cells, Cultured ,Innate immunity ,Invasive Pulmonary Aspergillosis ,Mice, Knockout ,Multidisciplinary ,Innate immune system ,biology ,Genetic Variation ,General Chemistry ,biology.organism_classification ,eye diseases ,Immunity, Innate ,3. Good health ,Complement system ,Transplantation ,Innate immune cells ,Mice, Inbred C57BL ,Serum Amyloid P-Component ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Pathogens - Abstract
© The Author(s) 2021. Open Access. This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/., Serum amyloid P component (SAP, also known as Pentraxin 2; APCS gene) is a component of the humoral arm of innate immunity involved in resistance to bacterial infection and regulation of tissue remodeling. Here we investigate the role of SAP in antifungal resistance. Apcs-/- mice show enhanced susceptibility to A. fumigatus infection. Murine and human SAP bound conidia, activate the complement cascade and enhance phagocytosis by neutrophils. Apcs-/- mice are defective in vivo in terms of recruitment of neutrophils and phagocytosis in the lungs. Opsonic activity of SAP is dependent on the classical pathway of complement activation. In immunosuppressed mice, SAP administration protects hosts against A. fumigatus infection and death. In the context of a study of hematopoietic stem-cell transplantation, genetic variation in the human APCS gene is associated with susceptibility to invasive pulmonary aspergillosis. Thus, SAP is a fluid phase pattern recognition molecule essential for resistance against A. fumigatus., The contribution of the European Commission (ERC project PHII-669415; FP7 project 281608 TIMER; ESA/ITN, H2020-MSCA-ITN-2015-676129), Ministero dell’Istruzione, dell’Università e della Ricerca (MIUR) (project FIRB RBAP11H2R9), Associazione Italiana Ricerca sul Cancro (AIRC IG-19014 and IG-21714, AIRC 5 × 1000 −9962 and −21147), the Italian Ministry of Health, the Northern Portugal Regional Operational Programme (NORTE 2020), under the Portugal 2020 Partnership Agreement, through the European Regional Development Fund (ERDF) (NORTE-01-0145-FEDER-000013 and NORTE-01-0145-FEDER-000023), the Fundação para a Ciência e Tecnologia (FCT) (UIDB/50026/2020, UIDP/50026/2020, PTDC/SAU-SER/29635/2017, PTDC/MED-GEN/28778/2017, CEECIND/04058/2018 and CEECIND/03628/2017), the European Union’s Horizon 2020 research and innovation program under grant agreement no. 847507 and the “la Caixa” Foundation (ID 100010434) and FCT under the agreement LCF/PR/HR17/52190003 is gratefully acknowledged.
- Published
- 2021
30. Controlled Infection of Humans with the Hookworm Parasite Necator americanus to Accelerate Vaccine Development
- Author
-
Rodrigo Correa-Oliveira, David I. Pritchard, Jeffrey M. Bethony, Maria Elena Bottazzi, David Diemert, and John M. Hawdon
- Subjects
Hookworm vaccine ,biology ,business.industry ,Endemic area ,biology.organism_classification ,Necator americanus ,Vaccination ,Clinical trial ,parasitic diseases ,Immunology ,Medicine ,Parasite hosting ,Clinical efficacy ,business ,Hookworm infection ,medicine.drug - Abstract
In this chapter, we describe the scientific, technical, clinical and regulatory aspects of establishing a controlled human hookworm infection (CHHI) model in non-endemic and endemic geographical regions, to facilitate a pathway towards accelerated vaccine development. The success achieved in establishing the CHHI platform specifically allows the Human Hookworm Vaccine Initiative (HHVI) to accelerate its progress by establishing a human hookworm vaccination/challenge model (HVCM) in a hookworm endemic area of Brazil. The HVCM will permit the rapid and robust determination of clinical efficacy in adults, allowing for early selection of the most efficacious human hookworm vaccine (HHV) candidate(s) to advance into later-stage pivotal paediatric clinical trials and reduce the overall number of participants required to assess efficacy (Diemert et al. 2018).
- Published
- 2021
- Full Text
- View/download PDF
31. Location and expression kinetics of Tc24 in different life stages of Trypanosoma cruzi
- Author
-
Cristina Poveda, Kathryn M. Jones, Maria Elena Bottazzi, Peter J. Hotez, Leroy Versteeg, Rakesh Adhikari, Edwin Tijhaar, Jeroen Pollet, and Maria Jose Villar-Mondragon
- Subjects
Protozoan Vaccines ,Life Cycles ,RC955-962 ,Parasitemia ,Protozoology ,Mice ,Medical Conditions ,Arctic medicine. Tropical medicine ,Medicine and Health Sciences ,Enzyme-Linked Immunoassays ,Protozoans ,Staining ,B-Lymphocytes ,Mice, Inbred BALB C ,biology ,Antibodies, Monoclonal ,Gene Expression Regulation, Developmental ,Eukaryota ,Specimen preparation and treatment ,Infectious Diseases ,Female ,Protozoan Life Cycles ,Antibody ,Public aspects of medicine ,RA1-1270 ,RNA, Protozoan ,Research Article ,Amastigotes ,Trypanosoma ,medicine.drug_class ,Trypanosoma cruzi ,Context (language use) ,Celbiologie en Immunologie ,Monoclonal antibody ,Microbiology ,Immunity ,Virology ,parasitic diseases ,medicine ,Parasitic Diseases ,Animals ,Humans ,Life Science ,Chagas Disease ,Amastigote ,Immunoassays ,Hybridomas ,Host Cells ,Public Health, Environmental and Occupational Health ,Organisms ,DAPI staining ,Biology and Life Sciences ,Trypomastigotes ,medicine.disease ,biology.organism_classification ,Parasitic Protozoans ,Research and analysis methods ,Cell Biology and Immunology ,Nuclear staining ,biology.protein ,Immunologic Techniques ,WIAS ,Viral Transmission and Infection ,Developmental Biology - Abstract
Tc24-C4, a modified recombinant flagellar calcium-binding protein of Trypanosoma cruzi, is under development as a therapeutic subunit vaccine candidate to prevent or delay progression of chronic Chagasic cardiomyopathy. When combined with Toll-like receptor agonists, Tc24-C4 immunization reduces parasitemia, parasites in cardiac tissue, and cardiac fibrosis and inflammation in animal models. To support further research on the vaccine candidate and its mechanism of action, murine monoclonal antibodies (mAbs) against Tc24-C4 were generated. Here, we report new findings made with mAb Tc24-C4/884 that detects Tc24-WT and Tc24-C4, as well as native Tc24 in T. cruzi on ELISA, western blots, and different imaging techniques. Surprisingly, detection of Tc24 by Tc24-C/884 in fixed T. cruzi trypomastigotes required permeabilization of the parasite, revealing that Tc24 is not exposed on the surface of T. cruzi, making a direct role of antibodies in the induced protection after Tc24-C4 immunization less likely. We further observed that after immunostaining T. cruzi–infected cells with mAb Tc24-C4/884, the expression of Tc24 decreases significantly when T. cruzi trypomastigotes enter host cells and transform into amastigotes. However, Tc24 is then upregulated in association with parasite flagellar growth linked to re-transformation into the trypomastigote form, prior to host cellular escape. These observations are discussed in the context of potential mechanisms of vaccine immunity., Author summary Chagas disease is a chronic infection with Trypanosoma cruzi (T. cruzi) that affects approximately 8 million people worldwide and may cause chronic heart inflammation. The vaccine candidate Tc24-C4 is a recombinant version of the Tc24 protein, which is a flagellar calcium-binding protein expressed by T. cruzi. While animal challenge studies have shown that targeting Tc24 is very promising, it is not fully understood how Tc24 is presented to the immune system. Here, we were able to localize Tc24 in flagellated T. cruzi parasites using a novel Tc24-specific monoclonal antibody. The results showed that Tc24 is not exposed on the outside of the parasite, which suggests that antibodies against Tc24 could not bind parasites during the infection. Then, by analyzing Tc24 expression in T. cruzi—infected host cells over time, we observed that Tc24 expression is reduced after the parasite enters the cells but is restored when parasites escape the host cell again. Our study provides more insights on the location and presence of Tc24 in T. cruzi during infection in the host, and we discuss our current understanding on the mechanisms of how the Tc24 vaccine may work.
- Published
- 2021
32. Control of Complement Activation by the Long Pentraxin PTX3: Implications in Age-Related Macular Degeneration
- Author
-
Matteo Stravalaci, Francesca Davi, Raffaella Parente, Marco Gobbi, Barbara Bottazzi, Alberto Mantovani, Anthony J. Day, Simon J. Clark, Mario R. Romano, and Antonio Inforzato
- Subjects
alternative pathway ,medicine ,Pharmacology (medical) ,pentraxin 3 ,age-related macular degeneration ,complement system ,Pharmacology ,pentraxins ,Retinal pigment epithelium ,Pentraxins ,biology ,Chemistry ,retinal pigmented epithelium ,lcsh:RM1-950 ,PTX3 ,Brief Research Report ,Macular degeneration ,medicine.disease ,vitreous humor ,eye diseases ,C3-convertase ,Cell biology ,Complement system ,lcsh:Therapeutics. Pharmacology ,medicine.anatomical_structure ,Factor H ,biology.protein ,Alternative complement pathway ,sense organs - Abstract
Dysregulation of the complement system is central to age-related macular degeneration (AMD), the leading cause of blindness in the developed world. Most of the genetic variation associated with AMD resides in complement genes, with the greatest risk associated with polymorphisms in the complement factor H (CFH) gene; factor H (FH) is the major inhibitor of the alternative pathway (AP) of complement that specifically targets C3b and the AP C3 convertase. Long pentraxin 3 (PTX3) is a soluble pattern recognition molecule that has been proposed to inhibit AP activation via recruitment of FH. Although present in the human retina, if and how PTX3 plays a role in AMD is still unclear. In this work we demonstrated the presence of PTX3 in the human vitreous and studied the PTX3-FH-C3b crosstalk and its effects on complement activation in a model of retinal pigment epithelium (RPE). RPE cells cultured in inflammatory AMD-like conditions overexpressed the PTX3 protein, and up-regulated AP activating genes. PTX3 bound RPE cells in a physiological setting, however this interaction was reduced in inflammatory conditions, whereby PTX3 had no complement-inhibiting activity on inflamed RPE. However, on non-cellular surfaces, PTX3 formed a stable ternary complex with FH and C3b that acted as a “hot spot” for complement inhibition. Our findings suggest a protective role for PTX3 in response to complement dysregulation in AMD and point to a novel mechanism of complement regulation by this pentraxin with potential implications in pathology and pharmacology of AMD.
- Published
- 2020
- Full Text
- View/download PDF
33. Macrophage expression and prognostic significance of the long pentraxin PTX3 in COVID-19
- Author
-
Giuseppe Gritti, Fabiano Di Marco, Michele Ciccarelli, Francesco Landi, Veronica Zanon, Marina Sironi, Monica Bacci, Alberto Mantovani, Claudio Angelini, Daniele Piovani, Alessandro Rambaldi, Maria De Santis, Marco Folci, Roberto Leone, Alessandro Protti, Sadaf Davoudian, Maurizio Cecconi, Andrea Gianatti, Clelia Peano, Domenico Supino, Ilaria My, Barbara Bottazzi, Enrico Brunetta, Federico Raimondi, Stefanos Bonovas, Cecilia Garlanda, Gianmarco Spata, Silvia Carnevale, and Sarah N. Mapelli
- Subjects
0301 basic medicine ,Adult ,Male ,Neutrophils ,Immunology ,Inflammation ,Peripheral blood mononuclear cell ,Monocytes ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Macrophage ,Humans ,Immunology and Allergy ,Epidemics ,Cells, Cultured ,A549 cell ,Innate immune system ,biology ,business.industry ,SARS-CoV-2 ,Gene Expression Profiling ,Macrophages ,C-reactive protein ,COVID-19 ,Endothelial Cells ,PTX3 ,Middle Aged ,Prognosis ,Serum Amyloid P-Component ,030104 developmental biology ,C-Reactive Protein ,A549 Cells ,biology.protein ,Immunohistochemistry ,Female ,medicine.symptom ,business ,030215 immunology - Abstract
Long pentraxin 3 (PTX3) is an essential component of humoral innate immunity, involved in resistance to selected pathogens and in the regulation of inflammation1-3. The present study was designed to assess the presence and significance of PTX3 in Coronavirus Disease 2019 (COVID-19)4-7. RNA-sequencing analysis of peripheral blood mononuclear cells, single-cell bioinformatics analysis and immunohistochemistry of lung autopsy samples revealed that myelomonocytic cells and endothelial cells express high levels of PTX3 in patients with COVID-19. Increased plasma concentrations of PTX3 were detected in 96 patients with COVID-19. PTX3 emerged as a strong independent predictor of 28-d mortality in multivariable analysis, better than conventional markers of inflammation, in hospitalized patients with COVID-19. The prognostic significance of PTX3 abundance for mortality was confirmed in a second independent cohort (54 patients). Thus, circulating and lung myelomonocytic cells and endothelial cells are a major source of PTX3, and PTX3 plasma concentration can serve as an independent strong prognostic indicator of short-term mortality in COVID-19.
- Published
- 2020
- Full Text
- View/download PDF
34. Genetic Modification to Design a Stable Yeast-expressed Recombinant SARS-CoV-2 Receptor Binding Domain as a COVID-19 Vaccine Candidate
- Author
-
Brian Keegan, Jungsoon Lee, Bin Zhan, Maria Jose Villar, Rakhi Tyagi Kundu, Cristina Poveda, Joanne Altieri Rivera, Jeroen Pollet, Leroy Versteeg, Maria Elena Bottazzi, Ana Carolina de Araujo Leao, Zhuyun Liu, Ulrich Strych, Junfei Wei, Wen-Hsiang Chen, Peter J. Hotez, Portia M. Gillespie, and Rakesh Adhikari
- Subjects
chemistry.chemical_classification ,Genetics ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Wild type ,Biology ,Protein tertiary structure ,Yeast ,law.invention ,Enzyme ,chemistry ,law ,Recombinant DNA ,Receptor ,Function (biology) - Abstract
BackgroundCoronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 has now spread worldwide to infect over 110 million people, with approximately 2.5 million reported deaths. A safe and effective vaccine remains urgently needed.MethodWe constructed three variants of the recombinant receptor-binding domain (RBD) of the SARS-CoV-2 spike (S) protein (residues 331-549) in yeast as follows: (1) a “wild type” RBD (RBD219-WT), (2) a deglycosylated form (RBD219-N1) by deleting the first N-glycosylation site, and (3) a combined deglycosylated and cysteine-mutagenized form (C538A-mutated variant (RBD219-N1C1)). We compared the expression yields, biophysical characteristics, and functionality of the proteins produced from these constructs.Results and conclusionsThese three recombinant RBDs showed similar secondary and tertiary structure thermal stability and had the same affinity to their receptor, angiotensin-converting enzyme 2 (ACE-2), suggesting that the selected deletion or mutations did not cause any significant structural changes or alteration of function. However, RBD219-N1C1 had a higher fermentation yield, was easier to purify, was not hyperglycosylated, and had a lower tendency to form oligomers, and thus was selected for further vaccine development and evaluation.General significanceBy genetic modification, we were able to design a better-controlled and more stable vaccine candidate, which is an essential and important criterion for any process and manufacturing of biologics or drugs for human use.
- Published
- 2020
- Full Text
- View/download PDF
35. Venous thromboembolism and COVID-19: a single center experience from an academic tertiary referral hospital of Northern Italy
- Author
-
Melazzini, Federica, Colaneri, Marta, Fumoso, Federica, Freddi, Giulia, Lenti, Marco Vincenzo, Pieri, Teresa Chiara, Piloni, Davide, Noris, Patrizia, Pieresca, Carla, Preti, Paola Stefania, Russo, Mariaconcetta, Corsico, Angelo, Tavazzi, Guido, Baldanti, Fausto, Triarico, Antonio, Mojoli, Francesco, Bruno, Raffaele, Di Sabatino, Antonio, Aronico, Nicola, Bergamaschi, Gaetano, Bertolino, Giampiera, Codega, Silvia, Costanzo, Filippo, Cresci, Roberto, Delliponti, Angela, Derosa, Giuseppe, Di Stefano, Michele, Falaschi, Francesco, Iadarola, Carmine, Lovati, Elisabetta, Lucotti, Pietro Carlo, Martignoni, Alessandra, Mengoli, Caterina, Miceli, Emanuela, Mugellini, Amedeo, Muggia, Chiara, Pagani, Elisabetta, Palumbo, Ilaria, Pecci, Alessandro, Perrone, Tiziano, Sgarlata, Carmelo, Siciliani, Luisa, Staniscia, Andrea, Vjera, Francesca Torello, Achilli, Giovanna, Agostinelli, Andrea, Antoci, Valentina, Ballesio, Alessia, Banfi, Francesco, Barteselli, Chiara, Benedetti, Irene, de Andreis, Federica Borrelli, Brattoli, Michele, Calabretta, Francesca, Cambiè, Ginevra, Canta, Roberta, Conca, Federico, Coppola, Luigi, Cremonte, Elisa Maria, Croce, Gabriele, Del Rio, Virginia, Di Terlizzi, Francesco, Ferrari, Maria Giovanna, Ferrari, Sara, Fiengo, Anna, Forni, Tommaso, Frigerio, Chiara, Fusco, Alessandra, Gabba, Margherita, Garolfi, Matteo, Gentile, Antonella, Gori, Giulia, Grandi, Giacomo, Grimaldi, Paolo, Lampugnani, Alice, Lapia, Francesco, Lepore, Federica, Lettieri, Gianluca, Mambella, Jacopo, Mercanti, Chiara, Merli, Stefania, Mordà, Francesco, Nardone, Alba, Pace, Luca, Padovini, Lucia, Parodi, Alessandro, Pellegrino, Ivan, Pitotti, Lavinia, Reduzzi, Margherita, Rigano, Giovanni, Romito, Giovanni, Rotola, Giorgio, Sabatini, Umberto, Salvi, Lucia, Santacroce, Giovanni, Savioli, Jessica, Soriano, Simone, Spataro, Carmine, Stefani, Debora, Aliberti, Anna Rita, Amatu, Alessandro, Anfossi, Laura, Arisi, Eric, Baldi, Chiara, Belliato, Mirko, Bellini, Lorenzo, Benzi, Alberto, Bichisao, Germana, Bolongaro, Antonia, Bottazzi, Andrea, Broglia, Federica, Bruschi, Giacomo, Caneva, Luca, Capaccio, Emanuele, Carboni, Valeria, Cavalloro, Fabrizio, Ciceri, Maria, Civardi, Luca, Delmonte, Maria Paola, Domenegati, Elisa Lucia, Ferrari, Federica, Ferrari, Fiorenza, Ferrari, Marta, Fuardo, Marinella, Gerletti, Maddalena Margherita, Gualdana, Simonetta, Ilardi, Marcella, Lo Coco, Claudia, Maggio, Giuseppe, Mascia, Maria Benedetta, Mencherini, Simonetta, Merati, Paola Maria, Mongodi, Silvia, Mori, Anna Maria, Morgante, Federica, Niebel, Thekla Larissa, Noli, Silvano, Orlando, Anita, Pagani, Michele, Passador, Debora, Pellicori, Simona, Perotti, Luciano, Picchioni, Raffaella, Poma, Silvia, Pozzi, Marco, Preti, Emanuela, Puce, Roberta, Radolovich, Danila Katia, Ragni, Gianluca, Repossi, Filippo, Riccardi, Francesca, Rizzardi, Roberto, Rodi, Giuseppe, Roldi, Emanuela, Rossi, Cristina, Sala Gallini, Giuseppe, Sciutti, Fabio, Sportiello, Debora, Ticozzelli, Giulia, Visconti, Federico, Zizzi, Silvia, Bagliani, Alessandro, Belotti, Corrado, Bossi, Chiara, Colombo, Andrea, Colombo, Costanza Natalia Julia, Cremascoli, Luca, Dammassa, Valentino, Discepoli, Roberto, Garlando, Maria Adelaide, Grandini, Filippo, Pellegrini, Andrea, Quaranta, Cecilia, Stella, Andrea, Torresani, Francesco, Mondelli, Mario, Brunetti, Enrico, Di Matteo, Angela, Seminari, Elena, Maiocchi, Laura, Zuccaro, Valentina, Pagnucco, Layla, Mariani, Bianca, Ludovisi, Serena, Lissandrin, Raffaella, Parisi, Aldo, Sacchi, Paolo, Patruno, Savino F. A., Michelone, Giuseppe, Gulminetti, Roberto, Zanaboni, Domenico, Novati, Stefano, Maserati, Renato, Orsolini, Paolo, Vecchia, Marco, Asperges, Erika, Di Filippo, Alessandro, Sambo, Margherita, Biscarini, Simona, Lupi, Matteo, Roda, Silvia, Gallazzi, Ilaria, Sachs, Michele, Valsecchi, Pietro, Ferrari, Alessandra, Bosio, Matteo, Cascina, Alessandro, Conio, Valentina, Di Domenica, Rita, Donnetta, Anna, Fraolini, Elia, Gualtieri, Giuseppe, Mangiarotti, Patrizia, Mariani, Francesca, Meloni, Federica, Oggionni, Tiberio, Pasturenzi, Lidia, Ronzoni, Vanessa, Saracino, Laura, Stella, Giulia, Tomaselli, Stefano, Abbate, Tommaso, Accordino, Giulia, Bertuccio, Francesco, Burattini, Cecilia, Cacciatore, Elisa, Cattaneo, Elena, Chino, Vittorio, Coretti, Manuela, Della Zoppa, Matteo, Infusino, Cristina, Lettieri, Sara, Maccabruni, Valeria, Mancinelli, Silvia, Tirelli, Claudio, and Vertui, Valentina
- Subjects
medicine.medical_specialty ,Coronavirus disease 2019 (COVID-19) ,030204 cardiovascular system & hematology ,Single Center ,Tertiary referral hospital ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Internal Medicine ,Medicine ,cardiovascular diseases ,Mean platelet volume ,biology ,SARS-CoV-2 ,business.industry ,Pulmonary embolism ,C-reactive protein ,Anticoagulants ,Thrombosis ,equipment and supplies ,medicine.disease ,Im - Original ,030220 oncology & carcinogenesis ,biology.protein ,Emergency Medicine ,Observational study ,business - Abstract
Preliminary evidence supports the notion that COVID-19 patients may have an increased susceptibility to develop venous thromboembolism (VTE). However, the magnitude of this association still needs to be defined. Furthermore, clinical predictors of thrombogenesis, and the relationship with the inflammatory status are currently unknown. On this basis, we conducted a retrospective, observational study on 259 consecutive COVID-19 patients admitted to an academic tertiary referral hospital in Northern Italy between March 19th and April 6th, 2020. Records of COVID-19 patients with a definite VTE event were reviewed for demographic information, co-morbidities, risk factors for VTE, laboratory tests, and anticoagulation treatment. Twenty-five cases among 259 COVID-19 patients developed VTE (9.6%), all of them having a Padua score > 4, although being under standard anticoagulation prophylaxis since hospital admission. In the VTE subcohort, we found a significant positive correlation between platelet count (PLT) and either C reactive protein (CRP) (p p = 0.0013), while a significant inverse correlation was observed between PLT and mean platelet volume (p p p = 0.002 and p = 0.005, respectively). No significant difference was found in d-dimer levels between VTE and non VTE patients, while significantly higher levels of LDH (p = 0.04) and IL-6 (p = 0.04) were observed in VTE patients in comparison to non-VTE patients. In conclusion, our findings showed a quite high prevalence of VTE in COVID-19 patients. Raised inflammatory indexes and increased serum levels of pro-inflammatory cytokines should raise the clinical suspicion of VTE.
- Published
- 2020
- Full Text
- View/download PDF
36. Repeat-Driven Generation of Antigenic Diversity in a Major Human Pathogen
- Author
-
Jaime A. Costales, José E. Calzada, Edmundo C. Grisard, Santuza M. R. Teixeira, João Luís Reis-Cunha, Kathryn M. Jones, Gabriel Machado Matos, Carlos Talavera-López, Peter J. Hotez, Michael A. Miles, Hernán J. Carrasco, Rodion Gorchakov, Mario J. Grijalva, Azael Saldaña, Felipe Guhl, Barbara A. Burleigh, Kristy O. Murray, Sofía Ocaña-Mayorga, Michael D. Lewis, Louisa A. Messenger, Daniella Castanheira Bartholomeu, Maria Elena Bottazzi, Matthew Yeo, Melissa S. Nolan, Juan David Ramírez, and Björn Andersson
- Subjects
0301 basic medicine ,Microbiology (medical) ,parasitology ,genome sequence ,Trypanosoma cruzi ,030231 tropical medicine ,Immunology ,lcsh:QR1-502 ,Sequence assembly ,Retrotransposon ,Biology ,Microbiology ,Genome ,Synteny ,lcsh:Microbiology ,pathology of infectious diseases ,03 medical and health sciences ,Cellular and Infection Microbiology ,0302 clinical medicine ,parasitic diseases ,Antigenic variation ,Gene ,Original Research ,Genetics ,Whole genome sequencing ,population genetics ,microbial genomics ,biology.organism_classification ,Antigenic Variation ,030104 developmental biology ,Infectious Diseases ,Multigene Family ,tropical medicine - Abstract
Trypanosoma cruzi, a zoonotic kinetoplastid protozoan parasite, is the causative agent of American trypanosomiasis (Chagas disease). Having a very plastic, repetitive and complex genome, the parasite displays a highly diverse repertoire of surface molecules, with pivotal roles in cell invasion, immune evasion and pathogenesis. Before 2016, the complexity of the genomic regions containing these genes impaired the assembly of a genome at chromosomal level, making it impossible to study the structure and function of the several thousand repetitive genes encoding the surface molecules of the parasite. We here describe the genome assembly of the Sylvio X10/1 genome sequence, which since 2016 has been used as a reference genome sequence for T. cruzi clade I (TcI), produced using high coverage PacBio single-molecule sequencing. It was used to analyze deep Illumina sequence data from 34 T. cruzi TcI isolates and clones from different geographic locations, sample sources and clinical outcomes. Resolution of the surface molecule gene distribution showed the unusual duality in the organization of the parasite genome, a synteny of the core genomic region with related protozoa flanked by unique and highly plastic multigene family clusters encoding surface antigens. The presence of abundant interspersed retrotransposons in these multigene family clusters suggests that these elements are involved in a recombination mechanism for the generation of antigenic variation and evasion of the host immune response on these TcI strains. The comparative genomic analysis of the cohort of TcI strains revealed multiple cases of such recombination events involving surface molecule genes and has provided new insights into T. cruzi population structure.
- Published
- 2020
37. Alterations to the cardiac metabolome induced by chronicT. cruziinfection relate to the degree of cardiac pathology
- Author
-
Kathryn M. Jones, Ekram Hossain, Kristyn Hoffman, Zongyuan Liu, Peter J. Hotez, Laura-Isobel McCall, and Maria Elena Bottazzi
- Subjects
Pathology ,medicine.medical_specialty ,biology ,business.industry ,Cardiac fibrosis ,Inflammation ,medicine.disease ,biology.organism_classification ,Pathogenesis ,Fibrosis ,Pathognomonic ,Metabolome ,Medicine ,Biomarker (medicine) ,medicine.symptom ,business ,Trypanosoma cruzi - Abstract
Chronic Chagasic cardiomyopathy (CCC) is a Neglected Tropical Disease caused by the parasiteTrypanosoma cruzi. The pathognomonic findings in symptomatic CCC patients and animal models includes diffuse cardiac fibrosis and inflammation with persistent parasite presence in the heart. This study investigated chemical alterations in different regions of the heart in relation to cardiac pathology indicators to better understand the long-term pathogenesis of this neglected disease. We used data from echocardiography, fibrosis biomarkers, and histopathological analysis to fully evaluate cardiac pathology. Metabolites isolated from the pericardial and endocardial sides of the right ventricular myocardium were analyzed by liquid chromatography tandem mass spectrometry. The endocardial sections contained significantly less cardiac inflammation and fibrosis than the pericardial sections. Cardiac levels of acylcarnitines, phosphocholines, and other metabolites were significantly disrupted in accordance with cardiac fibrosis, inflammation, and serum fibrosis biomarker levels. These findings have potential implications in treatment and monitoring for CCC patients.
- Published
- 2020
- Full Text
- View/download PDF
38. Lancet COVID-19 Commission Statement on the occasion of the 75th session of the UN General Assembly
- Author
-
Vaira Vike-Freiberga, Min Zhu, Felipe Larraín Bascuñán, Chandrika Bahadur, Jong Koo Lee, Lan Xue, Lara B. Aknin, Vitor Gaspar, Gabriela Cuevas Barron, Andy Haines, Ismail Serageldin, Miriam Were, Peter J. Hotez, Maria Elena Bottazzi, Raj Shah, Salim S. Abdool Karim, María Fernanda Espinosa, Kirsten Brosbøl, Phoebe Koundouri, Muhammad Pate, Lauren Barredo, Guillaume Lafortune, Yanis Ben Amor, Alejandro Gaviria, Ozge Karadag Caman, Juliana G.E. Bartels, Srinath Reddy, Paul Polman, John Thwaites, Joseph G. Allen, Peter Daszak, Chen Wang, Ismini Ethridge, Jeffrey D. Sachs, and Emma Torres
- Subjects
medicine.medical_specialty ,United Nations ,Statement (logic) ,General assembly ,Advisory Committees ,Pneumonia, Viral ,MEDLINE ,Commission ,Betacoronavirus ,medicine ,Humans ,Session (computer science) ,Pandemics ,biology ,Viral Epidemiology ,business.industry ,SARS-CoV-2 ,COVID-19 ,General Medicine ,biology.organism_classification ,medicine.disease ,Commission Statement ,Pneumonia ,Family medicine ,Communicable Disease Control ,business ,Coronavirus Infections - Published
- 2020
39. Host Immunity and Inflammation to Pulmonary Helminth Infections
- Author
-
Jill E. Weatherhead, Pedro Gazzinelli-Guimaraes, John M. Knight, Ricardo Fujiwara, Peter J. Hotez, Maria Elena Bottazzi, and David B. Corry
- Subjects
0301 basic medicine ,Host immunity ,lcsh:Immunologic diseases. Allergy ,lung pathologies ,Lung Diseases, Parasitic ,Immunology ,Helminthiasis ,Inflammation ,Review ,Adaptive Immunity ,Immune tolerance ,Host-Parasite Interactions ,Immunomodulation ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Helminths ,parasitic diseases ,medicine ,Immunology and Allergy ,Animals ,Humans ,Schistosoma ,Life Cycle Stages ,biology ,Ascaris ,Host (biology) ,Immunity ,biology.organism_classification ,host immune response ,Immunity, Innate ,030104 developmental biology ,Organ Specificity ,inflammation ,Disease Susceptibility ,medicine.symptom ,hookworm ,lcsh:RC581-607 ,Biomarkers ,030215 immunology - Abstract
Helminths, including nematodes, cestodes and trematodes, are complex parasitic organisms that infect at least one billion people globally living in extreme poverty. Helminthic infections are associated with severe morbidity particularly in young children who often harbor the highest burden of disease. While each helminth species completes a distinct life cycle within the host, several helminths incite significant lung disease. This impact on the lungs occurs either directly from larval migration and host immune activation or indirectly from a systemic inflammatory immune response. The impact of helminths on the pulmonary immune response involves a sophisticated orchestration and activation of the host innate and adaptive immune cells. The consequences of activating pulmonary host immune responses are variable with several helminthic infections leading to severe, pulmonary compromise while others providing immune tolerance and protection against the development of pulmonary diseases. Further delineation of the convoluted interface between helminth infection and the pulmonary host immune responses is critical to the development of novel therapeutics that are critically needed to prevent the significant global morbidity caused by these parasites.
- Published
- 2020
40. Schistosoma haematobium Extracellular Vesicle Proteins Confer Protection in a Heterologous Model of Schistosomiasis
- Author
-
Luke Becker, Darren Pickering, Bemnet Amare Tedla, Javier Sotillo, Lei Wang, Maria Elena Bottazzi, Bin Zhan, Alex Loukas, Gebeyaw G Mekonnen, Mark S. Pearson, and National Health and Medical Research Council (Australia)
- Subjects
0301 basic medicine ,030231 tropical medicine ,Immunology ,Heterologous ,lcsh:Medicine ,Schistosomiasis ,Article ,Microbiology ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Tetraspanin ,law ,schistosomiasis ,vaccine ,Drug Discovery ,parasitic diseases ,medicine ,Pharmacology (medical) ,Pharmacology ,Schistosoma haematobium ,biology ,Vesicle ,lcsh:R ,Extracellular vesicle ,Extracellular vesicles ,biology.organism_classification ,medicine.disease ,030104 developmental biology ,Infectious Diseases ,tetraspanin ,biology.protein ,Recombinant DNA ,Antibody ,extracellular vesicles ,Vaccine - Abstract
Helminth parasites release extracellular vesicles which interact with the surrounding host tissues, mediating host&ndash, parasite communication and other fundamental processes of parasitism. As such, vesicle proteins present attractive targets for the development of novel intervention strategies to control these parasites and the diseases they cause. Herein, we describe the first proteomic analysis by LC-MS/MS of two types of extracellular vesicles (exosome-like, 120k pellet vesicles and microvesicle-like, 15k pellet vesicles) from adult Schistosoma haematobium worms. A total of 57 and 330 proteins were identified in the 120k pellet vesicles and larger 15k pellet vesicles, respectively, and some of the most abundant molecules included homologues of known helminth vaccine and diagnostic candidates such as Sm-TSP2, Sm23, glutathione S-transferase, saponins and aminopeptidases. Tetraspanins were highly represented in the analysis and found in both vesicle types. Vaccination of mice with recombinant versions of three of these tetraspanins induced protection in a heterologous challenge (S. mansoni) model of infection, resulting in significant reductions (averaged across two independent trials) in liver (47%, 38% and 41%) and intestinal (47%, 45% and 41%) egg burdens. These findings offer insight into the mechanisms by which anti-tetraspanin antibodies confer protection and highlight the potential that extracellular vesicle surface proteins offer as anti-helminth vaccines.
- Published
- 2020
41. Macrophage expression and prognostic significance of the long pentraxin PTX3 in COVID-19
- Author
-
Andrea Gianatti, Michele Ciccarelli, Gianmarco Spata, Sarah N. Mapelli, Maria De Santis, Roberto Leone, Alessandro Protti, Monica Bacci, Alberto Mantovani, Barbara Bottazzi, Marina Sironi, Enrico Brunetta, Marco Folci, Cecilia Garlanda, Claudio Angelini, Veronica Zanon, Ilaria My, and Maurizio Cecconi
- Subjects
medicine.medical_specialty ,Lung ,medicine.diagnostic_test ,biology ,business.industry ,Inflammation ,PTX3 ,Gastroenterology ,Peripheral blood mononuclear cell ,Pathogenesis ,Ferritin ,Bronchoalveolar lavage ,medicine.anatomical_structure ,Internal medicine ,medicine ,biology.protein ,Respiratory system ,medicine.symptom ,business - Abstract
PTX3 is an essential component of humoral innate immunity, involved in resistance to selected pathogens and in the regulation of inflammation. PTX3 plasma levels are associated with poor outcome in systemic inflammatory conditions and vascular pathology. The present study was designed to assess expression and significance of PTX3 in COVID-19. By bioinformatics analysis of public databases PTX3 expression was detected in lung respiratory cell lines exposed to SARS-CoV-2. By analysis at single cell level of COVID-19 circulating mononuclear cells, we found that PTX3 was selectively expressed by monocytes among circulating leukocytes. Moreover, in lung bronchoalveolar lavage fluid, single cell analysis revealed selective expression of PTX3 in neutrophils and macrophages, which play a major role in the pathogenesis of the disease. By immunohistochemistry, PTX3 was expressed by lung myelomocytic cells, type 2 pneumocytes and vascular endothelial cells. PTX3 plasma levels were determined by ELISA in 96 consecutive patients with a laboratory-confirmed diagnosis of COVID-19. Higher PTX3 plasma levels were observed in 52 (54.2%) patients admitted in ICU (median 21.0ng/mL, IQT 15.5-46.3 vs 12.4ng/mL IQT 6.1-20.2 in ward patients; p=0.0017) and in 22 (23%) patients died by 28 days (39.8ng/mL, IQT 20.2-75.7 vs 15.7ng/mL, IQT 8.2-21.6 in survivors; p=0.0001). After determining an optimal PTX3 cut-off for the primary outcome, the Kaplan-Meier curve showed an increased mortality in patients with PTX3>22.25ng/mL (Log-rank tests p22.25ng/mL showed an adjusted Hazard Ratio (aHR) of 7.6 (95%CI2.45-23.76) in predicting mortality. Performing a multivariate logistic regression including all inflammatory markers (PTX3, ferritin, D-Dimer, IL-6, and CRP), PTX3 was the only marker significantly associated with death (aHR 1.13;95%CI1.02-1.24; p=0.021). The results reported here suggest that circulating and lung myelomonocytic cells are a major source of PTX3 and that PTX3 plasma levels can serve as a strong prognostic indicator of short-term mortality in COVID-19.
- Published
- 2020
- Full Text
- View/download PDF
42. I Never Thought I Had 'Moral Courage,' until COVID-19 Happened
- Author
-
Maria Elena Bottazzi
- Subjects
medicine.medical_specialty ,Biomedical Research ,COVID-19 Vaccines ,media_common.quotation_subject ,Pneumonia, Viral ,Morals ,Betacoronavirus ,Stories from the Field ,Virology ,Pandemic ,medicine ,Humans ,Psychiatry ,Pandemics ,Courage ,media_common ,biology ,business.industry ,Personal narrative ,Viral Epidemiology ,SARS-CoV-2 ,Viral Vaccine ,Communication ,COVID-19 ,Viral Vaccines ,biology.organism_classification ,medicine.disease ,Moral courage ,Pneumonia ,Infectious Diseases ,Parasitology ,business ,Coronavirus Infections - Published
- 2020
43. Schistosoma haematobiumextracellular vesicle proteins confer protection in a heterologous model of schistosomiasis
- Author
-
Bin Zhan, Alex Loukas, Gebeyaw G Mekonnen, Javier Sotillo, Maria Elena Bottazzi, Mark S. Pearson, Lei Wang, Luke Becker, Bemnet Amare Tedla, and Darren Pickering
- Subjects
Schistosoma haematobium ,biology ,Vesicle ,Heterologous ,Schistosomiasis ,Extracellular vesicle ,biology.organism_classification ,medicine.disease ,Microbiology ,law.invention ,law ,biology.protein ,Recombinant DNA ,medicine ,Helminths ,Antibody - Abstract
Helminth parasites release extracellular vesicles which interact with the surrounding host tissues, mediating host-parasite communication and other fundamental processes of parasitism. As such, vesicle proteins present attractive targets for the development of novel intervention strategies to control these parasites and the diseases they cause. Herein, we describe the first proteomic analysis by LC-MS/MS of two types of extracellular vesicles (exosome-like, 120k pellet vesicles and microvesicle-like, 15k pellet vesicles) from adultSchistosoma haematobiumworms. A total of 57 and 330 proteins were identified in the 120k pellet vesicles and larger 15k pellet vesicles, respectively, and some of the most abundant molecules included homologues of known helminth vaccine and diagnostic candidates such asSm-TSP2,Sm23, glutathione S-tranferase, saposins and aminopeptidases. Tetraspanins were highly represented in the analysis and found in both vesicle types. Vaccination of mice with recombinant versions of three of these tetraspanins induced protection in a heterologous challenge (S. mansoni) model of infection, resulting in significant reductions (averaged across two independent trials) in liver (47%, 38% and 41%) and intestinal (47%, 45% and 41%) egg burdens. These findings offer insight into the mechanisms by which anti-tetraspanin antibodies confer protection and highlight the potential that extracellular vesicle surface proteins offer as anti-helminth vaccines.
- Published
- 2020
- Full Text
- View/download PDF
44. Author response for 'TLR4 agonist protects against Trypanosoma cruzi acute lethal infection by decreasing cardiac parasite burdens'
- Author
-
L.E Villanueva‐Lizama, Brian Keegan, Maria Elena Bottazzi, J.V Cruz‐Chan, A Kendricks, K Hoffman, Peter J. Hotez, L Versteeg, F Gusovsky, J Pollet, C.F Teh‐Poot, and K.M Jones
- Subjects
Agonist ,biology ,medicine.drug_class ,Lethal infection ,TLR4 ,medicine ,Parasite hosting ,Trypanosoma cruzi ,biology.organism_classification ,Microbiology - Published
- 2020
- Full Text
- View/download PDF
45. COVID-19 vaccines: neutralizing antibodies and the alum advantage
- Author
-
Ulrich Strych, Maria Elena Bottazzi, David B. Corry, and Peter J. Hotez
- Subjects
0301 basic medicine ,History ,2019-20 coronavirus outbreak ,COVID-19 Vaccines ,Coronavirus disease 2019 (COVID-19) ,medicine.medical_treatment ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Education ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Adjuvants, Immunologic ,medicine ,Animals ,Humans ,Adjuvants ,biology ,SARS-CoV-2 ,business.industry ,Alum ,Viral Vaccine ,Comment ,Spike Protein ,Viral Vaccines ,respiratory system ,Antibodies, Neutralizing ,Macaca mulatta ,Virology ,respiratory tract diseases ,Computer Science Applications ,030104 developmental biology ,chemistry ,biology.protein ,Alum Compounds ,Antibody ,Coronavirus Infections ,business ,Adjuvant ,030215 immunology - Abstract
Achieving high levels of neutralizing antibodies to the spike protein of SARS-CoV-2 in a safe manner is likely to be crucial for an effective vaccine. Here, we propose that aluminium-based adjuvants might hold the key to this., Here, Peter Hotez and colleagues discuss the advantages of using an aluminium-based adjuvant in candidate COVID-19 vaccines.
- Published
- 2020
- Full Text
- View/download PDF
46. A scalable and reproducible manufacturing process for Phlebotomus papatasi salivary protein PpSP15, a vaccine candidate for leishmaniasis
- Author
-
Wen-Hsiang Chen, Surafel Damena, Mun Peak Nyon, Peter J. Hotez, Zhuyun Liu, Amadeo B. Biter, Bin Zhan, Maria Elena Bottazzi, Rakhi Tyagi Kundu, and Ulrich Strych
- Subjects
0106 biological sciences ,Genetic Vectors ,Gene Expression ,Leishmaniasis, Cutaneous ,01 natural sciences ,law.invention ,Pichia pastoris ,03 medical and health sciences ,Cutaneous leishmaniasis ,law ,Immunity ,010608 biotechnology ,medicine ,Animals ,Humans ,Cloning, Molecular ,Salivary Proteins and Peptides ,Leishmaniasis Vaccines ,030304 developmental biology ,Leishmania ,0303 health sciences ,biology ,Protein Stability ,Leishmaniasis ,biology.organism_classification ,medicine.disease ,Virology ,Recombinant Proteins ,Sandfly ,Molecular Weight ,Phlebotomus ,Fermentation ,Saccharomycetales ,Recombinant DNA ,Insect Proteins ,Female ,Cell bank ,Biotechnology - Abstract
Cutaneous leishmaniasis is a parasitic and neglected tropical disease transmitted by the bites of sandflies. The emergence of cutaneous leishmaniasis in areas of war, conflict, political instability, and climate change has prompted efforts to develop a preventive vaccine. One vaccine candidate antigen is PpSP15, a 15 kDa salivary antigen from the sandfly Phlebotomus papatasi that facilitates the infection of the Leishmania parasite and has been shown to induce parasite-specific cell-mediated immunity. Previously, we developed a fermentation process for producing recombinant PpSP15 in Pichia pastoris and a two-chromatographic-step purification process at 100 mL scale. Here we expand the process design to the 10 L scale and examine its reproducibility by performing three identical process runs, an essential transition step towards technology transfer for pilot manufacture. The process was able to reproducibly recover 81% of PpSP15 recombinant protein with a yield of 0.75 g/L of fermentation supernatant, a purity level of 97% and with low variance among runs. Additionally, a freeze-thaw stability study indicated that the PpSP15 recombinant protein remains stable after undergoing three freeze-thaw cycles, and an accelerated stability study confirmed its stability at 37 °C for at least one month. A research cell bank for the expression of PpSP15 was generated and fully characterized. Collectively, the cell bank and the production process are ready for technology transfer for future cGMP pilot manufacturing.
- Published
- 2020
47. Safety and immunogenicity of a recombinant vaccine against Trypanosoma cruzi in Rhesus macaques
- Author
-
Claudia Herrera, Amitinder Kaur, Eric Dumonteil, Preston A. Marx, Peter J. Hotez, Weihong Tu, Kelly Goff, Marissa Fahlberg, E Haupt, Jaime Ortega-López, and Maria Elena Bottazzi
- Subjects
Chagas disease ,Protozoan Vaccines ,Trypanosoma cruzi ,030231 tropical medicine ,Antigens, Protozoan ,CD8-Positive T-Lymphocytes ,Peripheral blood mononuclear cell ,Article ,law.invention ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antigen ,law ,medicine ,Animals ,Chagas Disease ,030212 general & internal medicine ,Vaccines, Synthetic ,General Veterinary ,General Immunology and Microbiology ,biology ,Immunogenicity ,Public Health, Environmental and Occupational Health ,biology.organism_classification ,medicine.disease ,Virology ,Macaca mulatta ,Infectious Diseases ,Blood chemistry ,Recombinant DNA ,Molecular Medicine ,CD8 - Abstract
Chagas disease, caused by the protozoan parasite Trypanosoma cruzi is one of the most important neglected parasitic diseases in the Americas. Vaccines represent an attractive complementary or even an alternative for the control of T. cruzi infection and pre-clinical studies in mice demonstrated that trypomastigote surface antigen (TSA-1) and the flagellar calcium-binding (Tc24) parasite antigens are promising candidates for vaccine development. We performed here the first evaluation of the safety and immunogenicity of two recombinant vaccine antigens (named TSA1-C4 and Tc24-C4) in naïve non-human primates. Three rhesus macaques received 3 doses of each recombinant protein, formulated with E6020 (Eisai Co., Ltd.), a novel Toll-like receptor-4 agonist, in a stable emulsion. All parameters from blood chemistry and blood cell counts were stable over the course of the study and unaffected by the vaccine. A specific IgG response against both antigens was detectable after the first vaccine dose, and increased with the second dose. After three vaccine doses, stimulation of PBMCs with a peptide pool derived from TSA1-C4 resulted in the induction of TSA1-C4-specific TNFα-, IL-2-and IFNγ-producing CD4(+) in one or two animals while stimulation with a peptide pool derived from Tc24-C4 only activated IFNγ-producing CD4(+)T cells in one animal. In two animals there was also activation of TSA1-C4-specific IL2-producing CD8(+) T cells. This is the first report of the immunogenicity of T. cruzi-derived recombinant antigens formulated as an emulsion with a TLR4 agonist in a non-human primate model. Our results strongly support the need for further evaluation of the preventive efficacy of this type of vaccine in non-human primates and explore the effect of the vaccine in a therapeutic model of naturally-infected Chagasic non-human primates, which would strengthen the rationale for the clinical development as a human vaccine against Chagas disease.
- Published
- 2020
48. TLR4 agonist protects against Trypanosoma cruzi acute lethal infection by decreasing cardiac parasite burdens
- Author
-
Julio Vladimir Cruz-Chan, Fabian Gusovsky, Liliana Estefanía Villanueva-Lizama, Jeroen Pollet, Kathryn M. Jones, Kristyn Hoffman, Maria Elena Bottazzi, April Kendricks, Leroy Versteeg, Brian Keegan, Peter J. Hotez, and Christian Teh-Poot
- Subjects
0301 basic medicine ,Agonist ,medicine.drug_class ,medicine.medical_treatment ,Trypanosoma cruzi ,030231 tropical medicine ,Immunology ,Inflammation ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Antigen ,Fibrosis ,medicine ,Animals ,Chagas Disease ,Phospholipids ,Mice, Inbred BALB C ,biology ,Immunotherapy ,medicine.disease ,biology.organism_classification ,Toll-Like Receptor 4 ,030104 developmental biology ,Cytokines ,Parasitology ,Female ,medicine.symptom ,Adjuvant - Abstract
E6020 is a synthetic agonist of Toll-like receptor-4 (TLR4). The purpose of this study was to evaluate the effect of different doses of E6020-SE on Trypanosoma cruzi-specific immune responses and its ability to confer protection against acute lethal infection in mice. Forty female BALB/c were infected with 500 trypomastigotes of T cruzi H1 strain, divided into four groups (n = 10) and treated at 7- and 14-day post-infection (dpi) with different doses of E6020-SE or PBS (control). Survival was followed for 51 days, mice were euthanized and hearts were collected to evaluate parasite burden, inflammation and fibrosis. We found significantly higher survival and lower parasite burdens in mice injected with E6020-SE at all doses compared to the control group. However, E6020-SE treatment did not significantly reduce cardiac inflammation or fibrosis. On the other hand, E6020-SE modulated Th1 and Th2 cytokines, decreasing IFN-γ and IL-4 in a dose-dependent manner after stimulation with parasite antigens. We conclude that E6020-SE alone increased survival by decreasing cardiac parasite burdens in BALB/c mice acutely infected with T cruzi but failed to prevent cardiac damage. Our results suggest that for optimal protection, a vaccine antigen is necessary to balance and orient a protective immune response.
- Published
- 2020
49. The Factor H-Binding Site of CspZ as a Protective Target against Multistrain, Tick-Transmitted Lyme Disease
- Author
-
Patricia L. Lederman, Thomas M. Hart, Ilva Lieknina, Wen-Hsiang Chen, Peter Kraiczy, Utpal Pal, Ashley L. Marcinkiewicz, Yi-Pin Lin, Kaspars Tars, Jennifer L. Yates, Nicholas J. Mantis, Maria Elena Bottazzi, and Xiuli Yang
- Subjects
Immunology ,Tick ,Microbiology ,Antibodies ,Mice ,Ticks ,Lyme disease ,Bacterial Proteins ,Mutant protein ,Borrelia ,medicine ,Animals ,Humans ,Binding site ,Lyme Disease ,Mice, Inbred BALB C ,Mice, Inbred C3H ,Binding Sites ,biology ,Lyme Disease Vaccines ,Complement System Proteins ,biology.organism_classification ,medicine.disease ,Virology ,Vaccination ,Infectious Diseases ,Immunization ,Borrelia burgdorferi ,Complement Factor H ,Microbial Immunity and Vaccines ,biology.protein ,Female ,Parasitology ,Antibody - Abstract
The spirochete Borrelia burgdorferisensu lato is the causative agent of Lyme disease (LD). The spirochetes produce the CspZ protein that binds to a complement regulator, factor H (FH). Such binding downregulates activation of host complement to facilitate spirochete evasion of complement killing. However, vaccination with CspZ does not protect against LD infection. In this study, we demonstrated that immunization with CspZ-YA, a CspZ mutant protein with no FH-binding activity, protected mice from infection by several spirochete genotypes introduced via tick feeding. We found that the sera from CspZ-YA-vaccinated mice more efficiently eliminated spirochetes and blocked CspZ FH-binding activity than sera from CspZ-immunized mice. We also found that vaccination with CspZ, but not CspZ-YA, triggered the production of anti-FH antibodies, justifying CspZ-YA as an LD vaccine candidate. The mechanistic and efficacy information derived from this study provides insights into the development of a CspZ-based LD vaccine.
- Published
- 2020
- Full Text
- View/download PDF
50. Potential for developing a SARS-CoV receptor-binding domain (RBD) recombinant protein as a heterologous human vaccine against coronavirus infectious disease (COVID)-19
- Author
-
Wen-Hsiang Chen, Maria Elena Bottazzi, and Peter J. Hotez
- Subjects
COVID-19 Vaccines ,medicine.drug_class ,viruses ,030231 tropical medicine ,Protein domain ,Pneumonia, Viral ,Immunology ,coronavirus ,Heterologous ,COVID-19 ,Heterologous vaccine ,subunit vaccine ,SARS ,SARS-CoV-2 ,receptor-binding domain ,Biology ,Monoclonal antibody ,medicine.disease_cause ,Virus ,law.invention ,03 medical and health sciences ,Betacoronavirus ,0302 clinical medicine ,Protein Domains ,Viral Envelope Proteins ,law ,medicine ,Humans ,Immunology and Allergy ,030212 general & internal medicine ,skin and connective tissue diseases ,Pandemics ,Coronavirus ,Pharmacology ,fungi ,virus diseases ,Viral Vaccines ,Virology ,Recombinant Proteins ,body regions ,Protein Subunits ,Recombinant DNA ,biology.protein ,Commentary ,Antibody ,Coronavirus Infections ,Article Commentary - Abstract
A SARS-CoV receptor-binding domain (RBD) recombinant protein was developed and manufactured under current good manufacturing practices in 2016. The protein, known as RBD219-N1 when formulated on Alhydrogel®, induced high-level neutralizing antibodies and protective immunity with minimal immunopathology in mice after a homologous virus challenge with SARS-CoV (MA15 strain). We examined published evidence in support of whether the SARS-CoV RBD219-N1 could be repurposed as a heterologous vaccine against Coronavirus Infectious Disease (COVID)-19. Our findings include evidence that convalescent serum from SARS-CoV patients can neutralize SARS-CoV-2. Additionally, a review of published studies using monoclonal antibodies (mAbs) raised against SARS-CoV RBD and that neutralizes the SARS-CoV virus in vitro finds that some of these mAbs bind to the receptor-binding motif (RBM) within the RBD, while others bind to domains outside this region within RBD. This information is relevant and supports the possibility of developing a heterologous SARS-CoV RBD vaccine against COVID-19, especially due to the finding that the overall high amino acid similarity (82%) between SARS-CoV and SARS-CoV-2 spike and RBD domains is not reflected in RBM amino acid similarity (59%). However, the high sequence similarity (94%) in the region outside of RBM offers the potential of conserved neutralizing epitopes between both viruses.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.