1
|
Sangeetha Vijayan P, Xavier J, Valappil MP. A review of immune modulators and immunotherapy in infectious diseases. Mol Cell Biochem 2024; 479:1937-1955. [PMID: 37682390 DOI: 10.1007/s11010-023-04825-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/05/2023] [Indexed: 09/09/2023]
Abstract
The human immune system responds to harmful foreign invaders frequently encountered by the body and employs defense mechanisms to counteract such assaults. Various exogenous and endogenous factors play a prominent role in maintaining the balanced functioning of the immune system, which can result in immune suppression or immune stimulation. With the advent of different immune-modulatory agents, immune responses can be modulated or regulated to control infections and other health effects. Literature provides evidence on various immunomodulators from different sources and their role in modulating immune responses. Due to the limited efficacy of current drugs and the rise in drug resistance, there is a growing need for new therapies for infectious diseases. In this review, we aim to provide a comprehensive overview of different immune-modulating agents and immune therapies specifically focused on viral infectious diseases.
Collapse
Affiliation(s)
- P Sangeetha Vijayan
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology [Govt. of India], Thiruvananthapuram, 695 012, Kerala, India
| | - Joseph Xavier
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology [Govt. of India], Thiruvananthapuram, 695 012, Kerala, India
| | - Mohanan Parayanthala Valappil
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology [Govt. of India], Thiruvananthapuram, 695 012, Kerala, India.
| |
Collapse
|
2
|
Chiappini E, Santamaria F, Marseglia GL, Marchisio P, Galli L, Cutrera R, de Martino M, Antonini S, Becherucci P, Biasci P, Bortone B, Bottero S, Caldarelli V, Cardinale F, Gattinara GC, Ciarcià M, Ciofi D, D'Elios S, Di Mauro G, Doria M, Indinnimeo L, Lo Vecchio A, Macrì F, Mattina R, Miniello VL, Del Giudice MM, Morbin G, Motisi MA, Novelli A, Palamara AT, Panatta ML, Pasinato A, Peroni D, Perruccio K, Piacentini G, Pifferi M, Pignataro L, Sitzia E, Tersigni C, Torretta S, Trambusti I, Trippella G, Valentini D, Valentini S, Varricchio A, Verga MC, Vicini C, Zecca M, Villani A. Prevention of recurrent respiratory infections : Inter-society Consensus. Ital J Pediatr 2021; 47:211. [PMID: 34696778 PMCID: PMC8543868 DOI: 10.1186/s13052-021-01150-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 09/09/2021] [Indexed: 12/12/2022] Open
Abstract
Recurrent respiratory infections (RRIs) are a common clinical condition in children, in fact about 25% of children under 1 year and 6% of children during the first 6 years of life have RRIs. In most cases, infections occur with mild clinical manifestations and the frequency of episodes tends to decrease over time with a complete resolution by 12 years of age. However, RRIs significantly reduce child and family quality of life and lead to significant medical and social costs.Despite the importance of this condition, there is currently no agreed definition of the term RRIs in the literature, especially concerning the frequency and type of infectious episodes to be considered. The aim of this consensus document is to propose an updated definition and provide recommendations with the intent of guiding the physician in the complex process of diagnosis, management and prevention of RRIs.
Collapse
Affiliation(s)
- Elena Chiappini
- Meyer University Hospital, Department of Health Science, University of Florence, Florence, Italy.
| | - Francesca Santamaria
- Departement of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Gian Luigi Marseglia
- Department of Pediatrics, University of Pavia IRCCS San Matteo foundation, Pavia, Italy
| | - Paola Marchisio
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luisa Galli
- Meyer University Hospital, Department of Health Science, University of Florence, Florence, Italy
| | - Renato Cutrera
- Respiratory Unit, Academic Pediatric Department, Pediatric Hospital Bambino Gesù IRCCS, Rome, Italy
| | - Maurizio de Martino
- Meyer University Hospital, Department of Health Science, University of Florence, Florence, Italy
| | - Sara Antonini
- Meyer University Hospital, Department of Health Science, University of Florence, Florence, Italy
| | - Paolo Becherucci
- Family Pediatrician, Local Health Unit, Lastra a Signa, Florence, Italy
| | - Paolo Biasci
- Family Pediatrician, Local Health Unit, Livorno, Italy
| | - Barbara Bortone
- Meyer University Hospital, Department of Health Science, University of Florence, Florence, Italy
| | - Sergio Bottero
- Airway Surgery Unit, Department of Pediatric Surgery, Bambino Gesù Children's Hospital, Rome, Italy
| | - Valeria Caldarelli
- Department of Obstetrics Gynaecology and Pediatrics, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Fabio Cardinale
- Department of Pediatrics, Pediatric Hospital Giovanni XXIII, University of Bari, Bari, Italy
| | | | - Martina Ciarcià
- Meyer University Hospital, Department of Health Science, University of Florence, Florence, Italy
| | - Daniele Ciofi
- Meyer University Hospital, Department of Health Science, University of Florence, Florence, Italy
| | - Sofia D'Elios
- Department of Clinical and Experimental Medicine, Section of Paediatrics, University of Pisa, Pisa, Italy
| | - Giuseppe Di Mauro
- Pediatric Primary Care, National Pediatric Health Care System, Caserta, Italy
| | - Mattia Doria
- Family Pediatrician, Local Health Unit, Chioggia, Venice, Italy
| | - Luciana Indinnimeo
- Pediatric Department "Sapienza", University of Rome, Policlinico Umberto I, Rome, Italy
| | - Andrea Lo Vecchio
- Departement of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Francesco Macrì
- Department of Pediatrics "Sapienza", University of Rome, Rome, Italy
| | - Roberto Mattina
- Department of Biomedical, Surgical, and Odontoiatric Sciences, Università degli Studi di Milano, Milan, Italy
| | | | - Michele Miraglia Del Giudice
- Department of Woman and Child and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Guido Morbin
- Family Pediatrician, Local Health Unit, Trento, Italy
| | - Marco Antonio Motisi
- Meyer University Hospital, Department of Health Science, University of Florence, Florence, Italy
| | - Andrea Novelli
- Meyer University Hospital, Department of Health Science, University of Florence, Florence, Italy
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Maria Laura Panatta
- Department of Otorhinolaryngology, IRCCS Bambino Gesù Pediatric Hospital, Rome, Italy
| | - Angela Pasinato
- Family Pediatrician, Local Health Unit, Torri di Quartesolo, Vicenza, Italy
| | - Diego Peroni
- Department of Clinical and Experimental Medicine, Section of Paediatrics, University of Pisa, Pisa, Italy
| | - Katia Perruccio
- Pediatric Oncology Hematology, Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Giorgio Piacentini
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, Pediatric Clinic, University of Verona, Verona, Italy
| | - Massimo Pifferi
- Department of Pediatrics Pulmonology and Allergology Section University Hospital of Pisa, Pisa, Italy
| | - Lorenzo Pignataro
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Emanuela Sitzia
- Department of Otorhinolaryngology, IRCCS Bambino Gesù Pediatric Hospital, Rome, Italy
| | - Chiara Tersigni
- Meyer University Hospital, Department of Health Science, University of Florence, Florence, Italy
| | - Sara Torretta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Irene Trambusti
- Department of Clinical and Experimental Medicine, Section of Paediatrics, University of Pisa, Pisa, Italy
| | - Giulia Trippella
- Meyer University Hospital, Department of Health Science, University of Florence, Florence, Italy
| | - Diletta Valentini
- Department of Pediatric and Infectious Disease Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Sandro Valentini
- Family Pediatrician, Local Health Unit, Colle Val d'Elsa, Siena, Italy
| | | | - Maria Carmen Verga
- Family Pediatrician, Local Health Unit Salerno, Vietri sul Mare, Salerno, Italy
| | - Claudio Vicini
- Department of Head-Neck Surgery, Otolaryngology, Head-Neck and Oral Surgery Unit, Morgagni Pierantoni Hospital, Forlì, Italy
| | - Marco Zecca
- Pediatric Hematology-Oncology, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Alberto Villani
- Department of Pediatric and Infectious Disease Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
3
|
Ivermectin presents effective and selective antileishmanial activity in vitro and in vivo against Leishmania infantum and is therapeutic against visceral leishmaniasis. Exp Parasitol 2020; 221:108059. [PMID: 33338468 DOI: 10.1016/j.exppara.2020.108059] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 12/03/2020] [Accepted: 12/13/2020] [Indexed: 12/19/2022]
Abstract
Treatment for visceral leishmaniasis (VL) is hindered mainly by the toxicity and/or high cost of therapeutic drugs. In addition, parasite resistance has been registered. Thus, there is an urgent need for the identification of novel, effective and low-cost antileishmanial agents. Since drug discovery is a long and expensive process, drug repositioning for treatment of leishmaniasis should be considered. In the present study, Ivermectin (IVE), a broad-spectrum drug used for treatment of parasitic diseases, was evaluated in vitro and in vivo against Leishmania infantum species. Results in vitro showed that IVE presented 50% Leishmania and macrophage inhibitory concentrations (IC50 and CC50, respectively) of 3.64 ± 0.48 μM and 427.50 ± 17.60 μM, respectively, with a selectivity index (SI) of 117.45; whereas Amphotericin B (AmpB), which was used as control, showed IC50 and CC50 values of 0.12 ± 0.05 μM and 1.06 ± 0.23 μM, respectively, with a corresponding SI of 8.90. Treatment with IVE effectively reduced the infection percentage and parasite burden in infected and treated macrophages and displayed a prophylactic activity by inhibiting macrophage infection with pre-treated parasites. Furthermore, preliminary studies suggested that IVE targets the parasite's mitochondria. Activity of IVE in its free format or incorporated into Pluronic® F127-based polymeric micelles (IVE/Mic) was also evaluated in vivo as a treating drug for L. infantum-infected BALB/c mice. Miltefosine was used as a control. Results showed that Miltefosine, IVE and IVE/Mic-treated animals presented significant reductions in the parasite load in their spleens, livers, bone marrows and draining lymph nodes, as well as development of an antileishmanial Th1-type immune response one and 15 days after treatment. Notably, IVE/Mic showed a better parasitological and immunological response in comparison to other alternative treatments. In conclusion, results suggest that IVE/Mic could be considered in future studies as a therapeutic alternative to treat VL.
Collapse
|
4
|
Lillo AM, Velappan N, Kelliher JM, Watts AJ, Merriman SP, Vuyisich G, Lilley LM, Coombs KE, Mastren T, Teshima M, Stein BW, Wagner GL, Iyer S, Bradbury ARM, Harris JF, Dichosa AE, Kozimor SA. Development of Anti- Yersinia pestis Human Antibodies with Features Required for Diagnostic and Therapeutic Applications. Immunotargets Ther 2020; 9:299-316. [PMID: 33294421 PMCID: PMC7716875 DOI: 10.2147/itt.s267077] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/16/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Yersinia pestis is a category A infective agent that causes bubonic, septicemic, and pneumonic plague. Notably, the acquisition of antimicrobial or multidrug resistance through natural or purposed means qualifies Y. pestis as a potential biothreat agent. Therefore, high-quality antibodies designed for accurate and sensitive Y. pestis diagnostics, and therapeutics potentiating or replacing traditional antibiotics are of utmost need for national security and public health preparedness. METHODS Here, we describe a set of human monoclonal immunoglobulins (IgG1s) targeting Y. pestis fraction 1 (F1) antigen, previously derived from in vitro evolution of a phage-display library of single-chain antibodies (scFv). We extensively characterized these antibodies and their effect on bacterial and mammalian cells via: ELISA, flow cytometry, mass spectrometry, spectroscopy, and various metabolic assays. RESULTS Two of our anti-F1 IgG (αF1Ig 2 and αF1Ig 8) stood out for high production yield, specificity, and stability. These two antibodies were additionally attractive in that they displayed picomolar affinity, did not compete when binding Y. pestis, and retained immunoreactivity upon chemical derivatization. Most importantly, these antibodies detected <1,000 Y. pestis cells in sandwich ELISA, did not harm respiratory epithelial cells, induced Y. pestis agglutination at low concentration (350 nM), and caused apparent reduction in cell growth when radiolabeled at a nonagglutinating concentration (34 nM). CONCLUSION These antibodies are amenable to the development of accurate and sensitive diagnostics and immuno/radioimmunotherapeutics.
Collapse
Affiliation(s)
- Antonietta M Lillo
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Nileena Velappan
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Julia M Kelliher
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Austin J Watts
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Samuel P Merriman
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Grace Vuyisich
- Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Laura M Lilley
- Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Kent E Coombs
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Tara Mastren
- Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Munehiro Teshima
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Benjamin W Stein
- Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Gregory L Wagner
- Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Srinivas Iyer
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | | | | | - Armand E Dichosa
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Stosh A Kozimor
- Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| |
Collapse
|
5
|
Soyer TG, Mendonça DVC, Tavares GSV, Lage DP, Dias DS, Ribeiro PAF, Perin L, Ludolf F, Coelho VTS, Ferreira ACG, Neves PHAS, Matos GF, Chávez-Fumagalli MA, Coimbra ES, Pereira GR, Coelho EAF, Antinarelli LMR. Evaluation of the in vitro and in vivo antileishmanial activity of a chloroquinolin derivative against Leishmania species capable of causing tegumentary and visceral leishmaniasis. Exp Parasitol 2019; 199:30-37. [PMID: 30817917 DOI: 10.1016/j.exppara.2019.02.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 02/09/2019] [Accepted: 02/23/2019] [Indexed: 12/23/2022]
Abstract
The treatment against leishmaniasis presents problems, since the currently used drugs are toxic and/or have high costs. In addition, parasite resistance has increased. As a consequence, in this study, a chloroquinolin derivative, namely 7-chloro-N,N-dimethylquinolin-4-amine or GF1059, was in vitro and in vivo tested against Leishmania parasites. Experiments were performed to evaluate in vitro antileishmanial activity and cytotoxicity, as well as the treatment of infected macrophages and the inhibition of infection using pre-treated parasites. This study also investigated the GF1059 mechanism of action in L. amazonensis. Results showed that the compound was highly effective against L. infantum and L. amazonensis, presenting a selectivity index of 154.6 and 86.4, respectively, against promastigotes and of 137.6 and 74.3, respectively, against amastigotes. GF1059 was also effective in the treatment of infected macrophages and inhibited the infection of these cells when parasites were pre-incubated with it. The molecule also induced changes in the parasites' mitochondrial membrane potential and cell integrity, and caused an increase in the reactive oxygen species production in L. amazonensis. Experiments performed in BALB/c mice, which had been previously infected with L. amazonensis promastigotes, and thus treated with GF1059, showed that these animals presented significant reductions in the parasite load when the infected tissue, spleen, liver, and draining lymph node were evaluated. GF1059-treated mice presented both lower parasitism and low levels of enzymatic markers, as compared to those receiving amphotericin B, which was used as control. In conclusion, data suggested that GF1059 can be considered a possible therapeutic target to be tested against leishmaniasis.
Collapse
Affiliation(s)
- Tauane G Soyer
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Débora V C Mendonça
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Grasiele S V Tavares
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniela P Lage
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniel S Dias
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Patrícia A F Ribeiro
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luisa Perin
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fernanda Ludolf
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vinicio T S Coelho
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Andreza C G Ferreira
- Pontifícia Universidade Católica de Minas Gerais, Departamento de Física e Química, Instituto de Ciências Exatas e Informática, Belo Horizonte, Minas Gerais, Brazil
| | - Pedro H A S Neves
- Pontifícia Universidade Católica de Minas Gerais, Departamento de Física e Química, Instituto de Ciências Exatas e Informática, Belo Horizonte, Minas Gerais, Brazil
| | - Guilherme F Matos
- Pontifícia Universidade Católica de Minas Gerais, Departamento de Física e Química, Instituto de Ciências Exatas e Informática, Belo Horizonte, Minas Gerais, Brazil
| | - Miguel A Chávez-Fumagalli
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Elaine S Coimbra
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Guilherme R Pereira
- Pontifícia Universidade Católica de Minas Gerais, Departamento de Física e Química, Instituto de Ciências Exatas e Informática, Belo Horizonte, Minas Gerais, Brazil
| | - Eduardo A F Coelho
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | - Luciana M R Antinarelli
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
6
|
Sharma C, Rokana N, Chandra M, Singh BP, Gulhane RD, Gill JPS, Ray P, Puniya AK, Panwar H. Antimicrobial Resistance: Its Surveillance, Impact, and Alternative Management Strategies in Dairy Animals. Front Vet Sci 2018; 4:237. [PMID: 29359135 PMCID: PMC5766636 DOI: 10.3389/fvets.2017.00237] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/15/2017] [Indexed: 01/10/2023] Open
Abstract
Antimicrobial resistance (AMR), one among the most common priority areas identified by both national and international agencies, is mushrooming as a silent pandemic. The advancement in public health care through introduction of antibiotics against infectious agents is now being threatened by global development of multidrug-resistant strains. These strains are product of both continuous evolution and un-checked antimicrobial usage (AMU). Though antibiotic application in livestock has largely contributed toward health and productivity, it has also played significant role in evolution of resistant strains. Although, a significant emphasis has been given to AMR in humans, trends in animals, on other hand, are not much emphasized. Dairy farming involves surplus use of antibiotics as prophylactic and growth promoting agents. This non-therapeutic application of antibiotics, their dosage, and withdrawal period needs to be re-evaluated and rationally defined. A dairy animal also poses a serious risk of transmission of resistant strains to humans and environment. Outlining the scope of the problem is necessary for formulating and monitoring an active response to AMR. Effective and commendably connected surveillance programs at multidisciplinary level can contribute to better understand and minimize the emergence of resistance. Besides, it requires a renewed emphasis on investments into research for finding alternate, safe, cost effective, and innovative strategies, parallel to discovery of new antibiotics. Nevertheless, numerous direct or indirect novel approaches based on host-microbial interaction and molecular mechanisms of pathogens are also being developed and corroborated by researchers to combat the threat of resistance. This review places a concerted effort to club the current outline of AMU and AMR in dairy animals; ongoing global surveillance and monitoring programs; its impact at animal human interface; and strategies for combating resistance with an extensive overview on possible alternates to current day antibiotics that could be implemented in livestock sector.
Collapse
Affiliation(s)
- Chetan Sharma
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Namita Rokana
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Mudit Chandra
- Department of Veterinary Microbiology, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Brij Pal Singh
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Rohini Devidas Gulhane
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Jatinder Paul Singh Gill
- School of Public Health and Zoonoses, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Pallab Ray
- Department of Medical Microbiology, Post Graduate Institute for Medical Education and Research (PGIMER), Chandigarh, India
| | - Anil Kumar Puniya
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| | - Harsh Panwar
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, India
| |
Collapse
|
7
|
Parihar SP, Hartley MA, Hurdayal R, Guler R, Brombacher F. Topical Simvastatin as Host-Directed Therapy against Severity of Cutaneous Leishmaniasis in Mice. Sci Rep 2016; 6:33458. [PMID: 27632901 PMCID: PMC5025842 DOI: 10.1038/srep33458] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/19/2016] [Indexed: 01/22/2023] Open
Abstract
We recently demonstrated that statins mediate protection against intracellular pathogens, Mycobacterium tuberculosis and Listeria monocytogenes in mice. Here, we investigated the immunomodulatory potential of simvastatin as a topical or systemic host-directed drug therapy in controlling inflammatory responses in an experimental mouse model of cutaneous leishmaniasis caused by Leishmania major (LV39). In an ear infection model, topical application of simvastatin directly on established lesions significantly reduced severity of the disease reflected by ear lesion size and ulceration. The host protective effect was further accompanied by decreased parasite burden in the ear and draining lymph nodes in both BALB/c and C57BL/6 mice. Pre-treatment of these mice on a low-fat cholesterol diet and systemic simvastatin also reduced footpad swelling, as well as parasite burdens and ulceration/necrosis in the more robust footpad infection model, demonstrating the prophylactic potential of simvastatin for cutaneous leishmaniasis. Mechanistically, following L. major infection, simvastatin-treated primary macrophages responded with significantly reduced cholesterol levels and increased production of hydrogen peroxide. Furthermore, simvastatin-treated macrophages displayed enhanced phagosome maturation, as revealed by increased LAMP-3 expression in fluorescent microscopy and Western blot analysis. These findings demonstrate that simvastatin treatment enhances host protection against L. major by increasing macrophage phagosome maturation and killing effector functions.
Collapse
Affiliation(s)
- Suraj P Parihar
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa.,Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa
| | - Mary-Anne Hartley
- Department of Biochemistry, University of Lausanne, Chemin des Boveresses 155, Epalinges, CH1066, Switzerland
| | - Ramona Hurdayal
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa.,Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa.,Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, Rondebosch-7701, Cape Town, South Africa
| | - Reto Guler
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa.,Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa.,Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa
| |
Collapse
|
8
|
Urt-Filho A, Oliveira RJ, Hermeto LC, Pesarini JR, David ND, Cantero WDB, Falcão G, Marks G, Antoniolli-Silva ACMB. Mesenchymal stem cell therapy promotes the improvement and recovery of renal function in a preclinical model. Genet Mol Biol 2016; 39:290-9. [PMID: 27275667 PMCID: PMC4910560 DOI: 10.1590/1678-4685-gmb-2015-0178] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 12/29/2015] [Indexed: 12/22/2022] Open
Abstract
Acute renal failure (ARF) is an extremely important public health issue in need of
novel therapies. The present study aimed to evaluate the capacity of mesenchymal stem
cell (MSC) therapy to promote the improvement and recovery of renal function in a
preclinical model. Wistar rats were used as the experimental model, and our results
show that cisplatin (5mg/kg) can efficiently induce ARF, as measured by changes in
biochemical (urea and creatinine) and histological parameters. MSC therapy performed
24h after the administration of chemotherapy resulted in normalized plasma urea and
creatinine levels 30 and 45d after the onset of kidney disease. Furthermore, MSC
therapy significantly reduced histological changes (intratubular cast formation in
protein overload nephropathy and tubular hydropic degeneration) in this ARF model.
Thus, considering that current therapies for ARF are merely palliative and that MSC
therapy can promote the improvement and recovery of renal function in this model
system, we suggest that innovative/alternative therapies involving MSCs should be
considered for clinical studies in humans to treat ARF.
Collapse
Affiliation(s)
- Antônio Urt-Filho
- Centro de Estudos em Células Tronco, Terapia Celular e Genética Toxicológica, Hospital Universitário "Maria Aparecida Pedrossian", Empresa Brasileira de Serviços Hospitalares, Campo Grande, MS, Brazil.,Programa de Pós-Graduação em Saúde e Desenvolvimento na Região Centro-Oeste, Faculdade de Medicina "Dr. Hélio Mandetta", Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Rodrigo Juliano Oliveira
- Centro de Estudos em Células Tronco, Terapia Celular e Genética Toxicológica, Hospital Universitário "Maria Aparecida Pedrossian", Empresa Brasileira de Serviços Hospitalares, Campo Grande, MS, Brazil.,Programa de Pós-Graduação em Saúde e Desenvolvimento na Região Centro-Oeste, Faculdade de Medicina "Dr. Hélio Mandetta", Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil.,Programa de Mestrado em Farmácia, Centro de Ciências Biológicas e da Saúde, Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Larissa Correa Hermeto
- Programa de Pós-Graduação em Clínica Veterinária, Faculdade de Ciências Agrária e Veterinária, Universidade Estadual Paulista "Júlio de Mesquita Filho", Jaboticabal, SP, Brazil
| | - João Renato Pesarini
- Centro de Estudos em Células Tronco, Terapia Celular e Genética Toxicológica, Hospital Universitário "Maria Aparecida Pedrossian", Empresa Brasileira de Serviços Hospitalares, Campo Grande, MS, Brazil.,Programa de Pós-Graduação em Saúde e Desenvolvimento na Região Centro-Oeste, Faculdade de Medicina "Dr. Hélio Mandetta", Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Natan de David
- Centro de Estudos em Células Tronco, Terapia Celular e Genética Toxicológica, Hospital Universitário "Maria Aparecida Pedrossian", Empresa Brasileira de Serviços Hospitalares, Campo Grande, MS, Brazil.,Programa de Mestrado em Farmácia, Centro de Ciências Biológicas e da Saúde, Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Wilson de Barros Cantero
- Centro de Estudos em Células Tronco, Terapia Celular e Genética Toxicológica, Hospital Universitário "Maria Aparecida Pedrossian", Empresa Brasileira de Serviços Hospitalares, Campo Grande, MS, Brazil.,Programa de Pós-Graduação em Saúde e Desenvolvimento na Região Centro-Oeste, Faculdade de Medicina "Dr. Hélio Mandetta", Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Gustavo Falcão
- Faculdade de Medicina "Dr. Hélio Mandetta", Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Guido Marks
- Programa de Pós-Graduação em Saúde e Desenvolvimento na Região Centro-Oeste, Faculdade de Medicina "Dr. Hélio Mandetta", Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Andréia Conceição Milan Brochado Antoniolli-Silva
- Centro de Estudos em Células Tronco, Terapia Celular e Genética Toxicológica, Hospital Universitário "Maria Aparecida Pedrossian", Empresa Brasileira de Serviços Hospitalares, Campo Grande, MS, Brazil.,Programa de Pós-Graduação em Saúde e Desenvolvimento na Região Centro-Oeste, Faculdade de Medicina "Dr. Hélio Mandetta", Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil
| |
Collapse
|
9
|
Targeting Immunomodulatory Agents to the Gut-Associated Lymphoid Tissue. NEURO-IMMUNO-GASTROENTEROLOGY 2016. [PMCID: PMC7123898 DOI: 10.1007/978-3-319-28609-9_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In addition to fluid haemostasis and lipid absorption, the lymphatic system and lymphoid tissues serve as the major host of immune cells where immune responses are evoked. Impaired function of the immune system might lead to serious diseases which are often treated by immunomodulators. This chapter briefly explores the physiology of an important part of the lymphatic system, the gut-associated lymphoid tissues (GALT). Currently used strategies for targeting GALT by immunomodulators for enhanced activity and/or decreased side effects are discussed. Strategies range from simple oral co-administration of immunomodulators with lipids to more advanced lipid-based formulations, polymer-based nanoparticle formulations and prodrugs. These targeting approaches successfully increase the concentration of immunomodulators achieved in the GALT and, more importantly, enhance immunomodulatory effects. Therefore, targeting immunomodulators to GALT represent a promising approach in the treatment of diseases where the immune system is actively involved.
Collapse
|
10
|
Martínez-Pérez JM, Robles-Pérez D, Benavides J, Morán L, Andrés S, Giráldez FJ, Rojo-Vázquez FA, Martínez-Valladares M. Effect of dietary supplementation with flaxseed oil or vitamin E on sheep experimentally infected with Fasciola hepatica. Res Vet Sci 2014; 97:71-9. [PMID: 24952681 DOI: 10.1016/j.rvsc.2014.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 05/07/2014] [Accepted: 05/25/2014] [Indexed: 01/10/2023]
Abstract
The effects of two diets supplemented with flaxseed oil (FO) or vitamin E (VE) were studied in sheep infected with Fasciola hepatica. The control group (CO), not supplemented, had higher weight gain and faecal egg count (P < 0.05) at the end of the study. At necropsy, the number (71.2 ± 26.5) and size of flukes were lower in the FO group (P < 0.01), probably due to the higher levels of white blood cells and lymphocytes (P < 0.01). This group also had higher red blood cell and haematocrit values (P < 0.01). The CO group had more severe hypoproteinaemia and hypoalbuminaemia, due to having the highest fluke burden (123.0 ± 35.2), and also the highest IgG1 titre (P < 0.01). The diet did not affect production or gene expression levels of IFN-γ and IL-4. On the other hand, the supplementation with VE led to a reduction in adult fluke burden (97.7 ± 39.9) and lower lipid oxidation in the liver (P < 0.001).
Collapse
Affiliation(s)
- J M Martínez-Pérez
- Departamento de Sanidad Animal, Parasitología y Enfermedades Parasitarias, Facultad de Veterinaria, Universidad de León, 24071 León, Spain
| | - D Robles-Pérez
- Departamento de Sanidad Animal, Parasitología y Enfermedades Parasitarias, Facultad de Veterinaria, Universidad de León, 24071 León, Spain
| | - J Benavides
- Instituto de Ganadería de Montaña (CSIC-ULE), Finca Marzanas, 24346 Grulleros (León), Spain
| | - L Morán
- Instituto de Ganadería de Montaña (CSIC-ULE), Finca Marzanas, 24346 Grulleros (León), Spain
| | - S Andrés
- Instituto de Ganadería de Montaña (CSIC-ULE), Finca Marzanas, 24346 Grulleros (León), Spain
| | - F J Giráldez
- Instituto de Ganadería de Montaña (CSIC-ULE), Finca Marzanas, 24346 Grulleros (León), Spain
| | - F A Rojo-Vázquez
- Departamento de Sanidad Animal, Parasitología y Enfermedades Parasitarias, Facultad de Veterinaria, Universidad de León, 24071 León, Spain; Instituto de Ganadería de Montaña (CSIC-ULE), Finca Marzanas, 24346 Grulleros (León), Spain
| | - M Martínez-Valladares
- Instituto de Ganadería de Montaña (CSIC-ULE), Finca Marzanas, 24346 Grulleros (León), Spain.
| |
Collapse
|
11
|
Cheng G, Hao H, Xie S, Wang X, Dai M, Huang L, Yuan Z. Antibiotic alternatives: the substitution of antibiotics in animal husbandry? Front Microbiol 2014; 5:217. [PMID: 24860564 PMCID: PMC4026712 DOI: 10.3389/fmicb.2014.00217] [Citation(s) in RCA: 340] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Accepted: 04/25/2014] [Indexed: 12/21/2022] Open
Abstract
It is a common practice for decades to use of sub-therapeutic dose of antibiotics in food-animal feeds to prevent animals from diseases and to improve production performance in modern animal husbandry. In the meantime, concerns over the increasing emergence of antibiotic-resistant bacteria due to the unreasonable use of antibiotics and an appearance of less novelty antibiotics have prompted efforts to develop so-called alternatives to antibiotics. Whether or not the alternatives could really replace antibiotics remains a controversial issue. This review summarizes recent development and perspectives of alternatives to antibiotics. The mechanism of actions, applications, and prospectives of the alternatives such as immunity modulating agents, bacteriophages and their lysins, antimicrobial peptides, pro-, pre-, and synbiotics, plant extracts, inhibitors targeting pathogenicity (bacterial quorum sensing, biofilm, and virulence), and feeding enzymes are thoroughly discussed. Lastly, the feasibility of alternatives to antibiotics is deeply analyzed. It is hard to conclude that the alternatives might substitute antibiotics in veterinary medicine in the foreseeable future. At the present time, prudent use of antibiotics and the establishment of scientific monitoring systems are the best and fastest way to limit the adverse effects of the abuse of antibiotics and to ensure the safety of animal-derived food and environment.
Collapse
Affiliation(s)
- Guyue Cheng
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University Wuhan, China
| | - Haihong Hao
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University Wuhan, China
| | - Shuyu Xie
- National Reference Laboratory of Veterinary Drug Residues, Huazhong Agricultural University Wuhan, China ; MOA Key Laboratory for the Detection of Veterinary Drug Residues in Foods, Huazhong Agricultural University Wuhan, China
| | - Xu Wang
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University Wuhan, China
| | - Menghong Dai
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University Wuhan, China
| | - Lingli Huang
- National Reference Laboratory of Veterinary Drug Residues, Huazhong Agricultural University Wuhan, China ; MOA Key Laboratory for the Detection of Veterinary Drug Residues in Foods, Huazhong Agricultural University Wuhan, China
| | - Zonghui Yuan
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University Wuhan, China ; National Reference Laboratory of Veterinary Drug Residues, Huazhong Agricultural University Wuhan, China ; MOA Key Laboratory for the Detection of Veterinary Drug Residues in Foods, Huazhong Agricultural University Wuhan, China
| |
Collapse
|
12
|
Bulgakova VA. [Immunomodulators for the prevention and treatment of acute respiratory infections: efficacy of azoximer bromide]. TERAPEVT ARKH 2014; 86:92-7. [PMID: 25804048 DOI: 10.17116/terarkh2014861292-97] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The review gives the results of a many-sided study of the basic properties of the therapeutic molecule of azoximer bromide (polyoxidonium). It analyzes our experience with the immunomodulator used to treat acute respiratory infections. Polyoxidonium has marked immunomodulatory (including interferon-producing) activity and a nonspecific protective effect against a broad range of pathogens, which is based on the regulation of systemic immunity rather than the direct inhibition of microorganisms. In addition, the drug has antitoxic and antioxidant properties. Azoximer bromide is used in combination with etiotropic therapy in the acute phase of disease and as monotherapy for rehabilitation or immunoprophylaxis of respiratory infections.
Collapse
|
13
|
Oliveira RJ, Salles MJS, da Silva AF, Kanno TYN, Lourenço ACDS, Leite VDS, Matiazi HJ, Pesarini JR, Ribeiro LR, Mantovani MS. In vivo evaluation of the antimutagenic and antigenotoxic effects of β-glucan extracted from Saccharomyces cerevisiae in acute treatment with multiple doses. Genet Mol Biol 2013; 36:413-24. [PMID: 24130450 PMCID: PMC3795161 DOI: 10.1590/s1415-47572013005000028] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 04/27/2013] [Indexed: 01/29/2023] Open
Abstract
Ample evidence suggests that cancer is triggered by mutagenic damage and diets or supplements capable of reducing such incidences can be related to the prevention of neoplasy development or to an improvement in life quality of patients who undergo chemotherapy. This research aimed to evaluate the antimutagenic and antigenotoxic activity of β-glucan. We set up 8 experimental groups: control (Group 1), cyclophosphamide (Group 2), Groups 3–5 to assess the effect of β-glucan administration, and Groups 6–8 to evaluate the association between cyclophosphamide and β-glucan. The intraperitonial concentrations of β-glucan used were 100, 150 and 200 mg/kg. Micronucleus and comet assays showed that within the first week of treatment β-glucan presented a damage reduction rate between 100–62.04% and 94.34–59.52% for mutagenic and genotoxic damages, respectively. This activity decreased as the treatment was extended. During the sixth week of treatment antimutagenicity rates were reduced to 59.51–39.83% and antigenotoxicity was not effective. This leads to the conclusion that the efficacy of β-glucan in preventing DNA damage is limited when treatment is extended, and that its use as a chemotherapeutic adjuvant need to be better clarified.
Collapse
Affiliation(s)
- Rodrigo Juliano Oliveira
- Centro de Estudos em Célula Tronco, Terapia Celular e Genética Toxicológica, Núcleo de Hospital Universitário, Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil . ; Programa de Pós-graduação em Saúde em Desenvolvimento na Região Centro-Oeste, Faculdade de Medicina "Dr. Hélio Mandetta", Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil . ; Programa de Mestrado em Farmácia, Centro de Ciências Biológicas e da Saúde, Universidade Federal de Mato Grosso do Sul, Campo Grande, MS, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Effect of lipopolysaccharide (LPS) from Ochrobactrum intermedium on sheep experimentally infected with Fasciola hepatica. Parasitol Res 2013; 112:2913-23. [DOI: 10.1007/s00436-013-3463-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 05/20/2013] [Indexed: 10/26/2022]
|
15
|
Abstract
The development of an infection involves interplay between the host's immune system and the virulence of the infecting microorganism. The traditional treatment of an infection involves antimicrobial chemotherapy to kill the organism. The use of immunotherapies in infections includes treatment options that modulate the immune response and can lead to control of infections. These therapies are expected to become more important therapeutic options with the increase in infections due to multidrug-resistant organisms and the increasing number of immunocompromised patients.
Collapse
Affiliation(s)
- Vivek Kak
- Allegiance Health, 1100 East Michigan Avenue, #305, Jackson, MI 49201, USA.
| | | | | | | |
Collapse
|
16
|
HANIEH H, NARABARA K, TANAKA Y, GU Z, ABE A, KONDO Y. Immunomodulatory effects of Alliums and Ipomoea batata extracts on lymphocytes and macrophages functions in White Leghorn chickens: In vitro study. Anim Sci J 2011; 83:68-76. [DOI: 10.1111/j.1740-0929.2011.00918.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
17
|
Giagulli C, Noerder M, Avolio M, Becker PD, Fiorentini S, Guzman CA, Caruso A. Pidotimod promotes functional maturation of dendritic cells and displays adjuvant properties at the nasal mucosa level. Int Immunopharmacol 2009; 9:1366-73. [PMID: 19712757 DOI: 10.1016/j.intimp.2009.08.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Revised: 08/07/2009] [Accepted: 08/10/2009] [Indexed: 10/20/2022]
Abstract
Mucosal dendritic cells (DCs) are very important in the process of antigen presentation to T cells, playing a key role in the induction of primary and secondary immune responses. Pidotimod is a synthetic substance capable of modulating immune cell functions, but the effect of pidotimod on human DCs has not been investigated yet. Here we demonstrate the ability of pidotimod to induce DC maturation and up-regulate the expression of HLA-DR and co-stimulatory molecules CD83 and CD86, which are fundamental for communication with adaptative immunity cells. Pidotimod also stimulated DCs to release high amounts of pro-inflammatory molecules such as MCP-1 and TNF-alpha cytokines and to drive T cell proliferation and differentiation towards a Th1 phenotype. Moreover, we demonstrate that pidotimod in vivo promotes strong and specific humoral and cellular immune response when co-administered intranasally with a model antigen. Taken together our data suggest the possibility to use pidotimod as adjuvant molecule to facilitate the activation of the innate immune system as well as to promote an effective mucosal and systemic immune response.
Collapse
Affiliation(s)
- Cinzia Giagulli
- Department of Experimental and Applied Medicine, Section of Microbiology, University of Brescia, Medical School, Brescia, Italy
| | | | | | | | | | | | | |
Collapse
|
18
|
In vitro Effects of Methanol Extracts of Korean Medicinal Fruits (Persimmon, Raspberry, Tomato) on Chicken Lymphocytes, Macrophages, and Tumor Cells. J Poult Sci 2009. [DOI: 10.2141/jpsa.46.149] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
19
|
Wang Z, Shao Y, Guo Y, Yuan J. Enhancement of Peripheral Blood CD8+T Cells and Classical Swine Fever Antibodies by Dietaryβ-1,3/1,6-glucan Supplementation in Weaned Piglets. Transbound Emerg Dis 2008; 55:369-76. [DOI: 10.1111/j.1865-1682.2008.01049.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
20
|
Mantovani MS, Bellini MF, Angeli JPF, Oliveira RJ, Silva AF, Ribeiro LR. beta-Glucans in promoting health: prevention against mutation and cancer. Mutat Res 2007; 658:154-61. [PMID: 17827055 DOI: 10.1016/j.mrrev.2007.07.002] [Citation(s) in RCA: 160] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2007] [Revised: 07/16/2007] [Accepted: 07/20/2007] [Indexed: 02/07/2023]
Abstract
The polysaccharides beta-glucans occur as a principal component of the cellular walls. Some microorganisms, such as yeast and mushrooms, and also cereals such as oats and barley, are of economic interest because they contain large amounts of beta-glucans. These substances stimulate the immune system, modulating humoral and cellular immunity, and thereby have beneficial effect in fighting infections (bacterial, viral, fungal and parasitic). beta-Glucans also exhibit hypocholesterolemic and anticoagulant properties. Recently, they have been demonstrated to be anti-cytotoxic, antimutagenic and anti-tumorogenic, making them promising candidate as pharmacological promoters of health.
Collapse
Affiliation(s)
- Mário S Mantovani
- Departamento de Biologia Geral, Universidade Estadual de Londrina, Londrina, PR, Brazil.
| | | | | | | | | | | |
Collapse
|
21
|
Lee SH, Lillehoj HS, Chun HK, Tuo W, Park HJ, Cho SM, Lee YM, Lillehoj EP. In vitro treatment of chicken peripheral blood lymphocytes, macrophages, and tumor cells with extracts of Korean medicinal plants. Nutr Res 2007; 27:362-366. [DOI: 10.1016/j.nutres.2007.04.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2006] [Revised: 03/16/2007] [Accepted: 04/10/2007] [Indexed: 11/30/2022]
|
22
|
Becker PD, Nörder M, Guzmán CA, Grinstein S. Immune modulator adamantylamide dipeptide stimulates efficient major histocompatibility complex class I-restricted responses in mice. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2007; 14:538-43. [PMID: 17344349 PMCID: PMC1865622 DOI: 10.1128/cvi.00316-06] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Adamantylamide L-alanyl-D-isoglutamine (AdDP) is a synthetic adjuvant which belongs to the family of the desmuramyl peptides. AdDP exerts its adjuvant properties when it is administered either by the parenteral or by the mucosal route, leading to the elicitation of strong humoral responses at both the systemic and the mucosal levels. However, very little is known about the effect of AdDP on cellular immunity. Here we demonstrate that AdDP is able to stimulate cellular responses, which are characterized by the release of gamma interferon by CD8+ T cells when they are restimulated with a major histocompatibility complex class I-restricted peptide and strong in vivo lymphocyte-mediated cytotoxic activity. The capacity of AdDP to stimulate the elicitation of both cellular and humoral adaptive responses makes this adjuvant a promising tool for the development of mucosal vaccine formulations.
Collapse
Affiliation(s)
- Pablo D Becker
- Department of Vaccinology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, D-38124 Braunschweig, Germany
| | | | | | | |
Collapse
|
23
|
Garay RP, Viens P, Bauer J, Normier G, Bardou M, Jeannin JF, Chiavaroli C. Cancer relapse under chemotherapy: why TLR2/4 receptor agonists can help. Eur J Pharmacol 2007; 563:1-17. [PMID: 17383632 DOI: 10.1016/j.ejphar.2007.02.018] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2006] [Revised: 02/07/2007] [Accepted: 02/08/2007] [Indexed: 01/16/2023]
Abstract
Liver or lung metastases usually relapse under chemotherapy. Such life-threatening condition urgently needs new, systemic anticancer compounds, with original and efficient mechanisms of action. In B16 melanoma mice treated with cyclophosphamide, D'Agostini et al. [D'Agostini, C., Pica, F., Febbraro, G., Grelli, S., Chiavaroli, C., Garaci, E., 2005. Antitumour effect of OM-174 and Cyclophosphamide on murine B16 melanoma in different experimental conditions. Int. Immunopharmacol. 5, 1205-1212.] recently found that OM-174, a chemically defined Toll-like receptor(TLR)2/4 agonist, reduces tumor progression and prolongs survival. Here we review 149 articles concerning molecular mechanisms of TLR2/4 agonists, alone or in combination with chemotherapy. It appears that TLR2/4 agonists induce a well controlled tumor necrosis factor-alpha (TNF-alpha) secretion, at plasma levels known to permeabilize neoangiogenic tumor vessels to the passage of cytotoxic drugs. Moreover, TLR2/4 agonists induce inducible nitric oxide synthase (iNOS) expression, and nitric oxide is able to induce apoptosis of chemotherapy-resistant tumor cell clones. Finally, TLR2/4-stimulation activates dendritic cell traffic and its associated tumor-specific, cytotoxic T-cell responses. Therefore, parenteral TLR2/4 agonists seem promising molecules to prolong survival in cancer patients who relapse under chemotherapy.
Collapse
|
24
|
Abstract
Recombinant interferon-alpha (IFN-alpha) was approved by regulatory agencies in many countries in 1986. As the first biotherapeutic approved, IFN-alpha paved the way for the development of many other cytokines and growth factors. Nevertheless, understanding the functions of the multitude of human IFNs and IFN-like cytokines has just touched the surface. This review summarizes the history of the purification of human IFNs and the key aspects of our current state of knowledge of human IFN genes, proteins, and receptors. All the known IFNs and IFN-like cytokines are described [IFN-alpha, IFN-beta, IFN-epsilon, IFN-kappa, IFN-omega, IFN-delta, IFN-tau, IFN-gamma, limitin, interleukin-28A (IL-28A), IL-28B, and IL-29] as well as their receptors and signal transduction pathways. The biological activities and clinical applications of the proteins are discussed. An extensive section on the evolution of these molecules provides some new insights into the development of these proteins as major elements of innate immunity. The overall structure of the IFNs is put into perspective in relation to their receptors and functions.
Collapse
Affiliation(s)
- Sidney Pestka
- Department of Molecular Genetics, Microbiology, and Immunology, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, Piscataway, NJ 08854-5635, USA.
| | | | | |
Collapse
|
25
|
Eberlin S, dos Santos LMB, Queiroz MLS. Uncaria tomentosa extract increases the number of myeloid progenitor cells in the bone marrow of mice infected with Listeria monocytogenes. Int Immunopharmacol 2005; 5:1235-46. [PMID: 15914328 DOI: 10.1016/j.intimp.2005.03.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2003] [Revised: 01/09/2004] [Accepted: 03/03/2005] [Indexed: 11/17/2022]
Abstract
In this study, we demonstrated that Uncaria tomentosa extract (UTE) protects mice from a lethal dose of Listeria monocytogenes when administered prophylactically at 50, 100, 150 and 200 mg/kg for 7 days, with survival rates up to 35%. These doses also prevented the myelosuppression and the splenomegaly caused by a sublethal infection with L. monocytogenes, due to increased numbers of granulocyte-macrophage progenitors (CFU-GM) in the bone marrow. Non-infected mice treated with 100 mg/kg UTE also presented higher numbers of CFU-GM in the bone marrow than the controls. Investigation of the production of colony-stimulating factors revealed increased colony-stimulating activity (CSA) in the serum of normal and infected mice pre-treated with UTE. Moreover, stimulation of myelopoiesis and CSA occurred in a dose-dependent manner, a plateaux being reached with the dose of 100 mg/kg. Further studies to investigate the levels of factors such as IL-1 and IL-6 were undertaken. We observed increases in the levels of IL-1 and IL-6 in mice infected with L. monocytogenes and treated with 100 mg/kg of UTE. White blood cells (WBC) and differential counting were also performed, and our results demonstrated no significant changes in these data, when infected mice were pre-treated with 100 mg/kg of UTE. All together, our results suggest that UTE indirectly modulates immune activity and probably disengages Listeria-induced supression of these responses by inducing a higher reserve of myeloid progenitors in the bone marrow in consequence of biologically active cytokine release (CSFs, IL-1 and IL-6).
Collapse
Affiliation(s)
- Samara Eberlin
- Departamento de Farmacologia/Hemocentro, Faculdade de Ciências Médicas, Universidade Estadual de Campinas (UNICAMP), C.P. 6111, CEP 13083-970, Campinas, SP, Brazil
| | | | | |
Collapse
|
26
|
Liu G, Zhai Q, Schaffner D, Popova T, Hayford A, Bailey C, Alibek K. Bacillus alcalophilus peptidoglycan induces IFN-alpha-mediated inhibition of vaccinia virus replication. ACTA ACUST UNITED AC 2004; 42:197-204. [PMID: 15364104 DOI: 10.1016/j.femsim.2004.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2003] [Revised: 04/16/2004] [Accepted: 05/10/2004] [Indexed: 01/12/2023]
Abstract
Bacterial products such as cell walls (CW) and peptidoglycan (PGN) are known to activate macrophages and NK cells during microbial infections. In this report, we demonstrated that whole CW and PGN of four Gram-positive bacteria are capable of enhancing the anti-poxviral activity of murine macrophage RAW 264.7 cells. Among the major Bacillus alcalophilus CW components, PGN contributes the most to antiviral activity and induces remarkably higher levels of IFN-alpha. Anti-IFN-alpha/beta antibody, but not anti-IFN-gamma, anti-IFN-gamma receptor, or anti-IL-12, reversed the PGN-induced inhibition of vaccinia virus replication and reduced nitric oxide (NO) production. Our data thus suggest that PGN induce antiviral activity through IFN-alpha and to a lesser extent, through NO production.
Collapse
Affiliation(s)
- Ge Liu
- Advanced Biosystems, Inc., George Mason University, 10900 University Blvd., MSN 1A8 Manassas, VA 20110, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Sharma P, Singh N, Garg R, Haq W, Dube A. Efficacy of human beta-casein fragment (54-59) and its synthetic analogue compound 89/215 against Leishmania donovani in hamsters. Peptides 2004; 25:1873-81. [PMID: 15501518 DOI: 10.1016/j.peptides.2004.06.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2003] [Revised: 06/15/2004] [Accepted: 06/17/2004] [Indexed: 12/15/2022]
Abstract
The characteristic feature of visceral leishmaniasis (VL) is the profound impairment of immune system of the infected host, which contributes significantly to the partial success of antileishmanial chemotherapy. Since in VL, cure is the combinatorial effect of drug and immune status of the host, the rationale approach towards antileishmanial chemotherapy would be to potentiate the immune functioning of the host to extract desired results. Towards this direction several rationally designed analogues of human beta-casein fragment (54-59) were evaluated for their ability to stimulate the non-specific resistance in hamsters against Leishmania donovani infection. By virtue of being derived from the food protein casein derivatives may be devoid of unwanted side effects associated with the substances of microbial origin, e.g. muramyl dipeptide (MDP). Out of this one peptide Val-Glu-Gly-Ile-Pro-Tyr (compound 89/215) had been reported to have such activity. In this communication, the prophylactic and therapeutic efficacy of the peptide along with its natural sequence has been evaluated in detail against experimental VL in hamsters. Their use as an adjunct to chemotherapy was also explored. Human beta-casein fragment, compound 89/215 and MDP were tested in vivo at various dose levels wherein compound 89/215 showed superiority over MDP at 3 mg/kg x 2 given intraperitoneally (i.p.). Compound 89/215 sensitized peritoneal macrophages acquired considerable resistance and only 24% of the cells were found infected in comparison to control peritoneal macrophages where 76.4% of the cells were found infected. Similarly, the efficacy of sodium antimony gluconate (SAG) in hamsters pretreated with compound 89/215 enhanced significantly (P < 0.001). This peptide also exhibited considerably good therapeutic efficacy when evaluated either alone or in combination with SAG in established infection of L. donovani.
Collapse
Affiliation(s)
- Preeti Sharma
- Division of Parasitology, Central Drug Research Institute, Post Box No. 173, Lucknow 226001, India
| | | | | | | | | |
Collapse
|
28
|
Pelizon AC, Kaneno R, Soares AM, Meira DA, Sartori A. Down-modulation of lymphoproliferation and interferon-gamma production by beta-glucan derived from Saccharomyces cerevisiae. Mem Inst Oswaldo Cruz 2004; 98:1083-7. [PMID: 15049094 DOI: 10.1590/s0074-02762003000800019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
beta-glucan, one of the major cell wall components of Saccharomyces cerevisiae, has been found to enhance immune functions. This study investigated in vivo and in vitro effects of beta-glucan on lymphoproliferation and interferon-gamma (IFN-gamma) production by splenic cells from C57BL/6 female mice. All experiments were performed with particulate beta-glucan derived from S. cerevisiae. Data demonstrated that both, i.p. administration of particulate beta-glucan (20 or 100 micrograms/animal) and in vitro stimulation of splenic cells (20 or 100 micrograms/ml of culture) decreased lymphoproliferation and IFN-gamma production induced by concanavalin A. These results suggest that beta-glucan can trigger a down-modulatory effect regulating a deleterious immune system hyperactivity in the presence of a strong stimulus.
Collapse
Affiliation(s)
- A C Pelizon
- Departamento de Microbiologia e Imunologia, Instituto de Biociências da Unesp, Faculdade de Medicina, Campus de Botucatu, 18618-000 Botucatu, SP, Brasil.
| | | | | | | | | |
Collapse
|
29
|
Abstract
Combating pathogenic organisms by combinatorial approaches involving appropriate immune response molecules and antimicrobial drugs represents a progessively more apparent and successful therapeutic paradigm for the treatment of acute and chronic persistent infectious diseases. This review explores areas of current innovation and provides an update of the present state of knowledge concerning combination of chemotherapy with several immune-based interventions in infections. In the future, a better understanding of microbial immune modulation and evasion may continue to open new avenues of inquiry and carefully targeted application of adjunctive immunotherapies.
Collapse
Affiliation(s)
- Hartmut Hengel
- Robert Koch Institute, Nordufer 20, D-13353 Berlin, Germany
| | | |
Collapse
|
30
|
Affiliation(s)
- K Noel Masihi
- Robert Koch Institute, Nordufer 20, 13353 Berlin, Germany.
| |
Collapse
|
31
|
de Waard R, Claassen E, Bokken GCAM, Buiting B, Garssen J, Vos JG. Enhanced immunological memory responses to Listeria monocytogenes in rodents, as measured by delayed-type hypersensitivity (DTH), adoptive transfer of DTH, and protective immunity, following Lactobacillus casei Shirota ingestion. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2003; 10:59-65. [PMID: 12522040 PMCID: PMC145274 DOI: 10.1128/cdli.10.1.59-65.2003] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2002] [Revised: 07/22/2002] [Accepted: 11/01/2002] [Indexed: 11/20/2022]
Abstract
We have investigated the effect of orally administered Lactobacillus casei Shirota (L. casei) on immunological memory, as measured by delayed-type hypersensitivity (DTH) and acquired cellular resistance (ACR). The studies were performed in animal models in which the animals were rendered immune by a primary Listeria monocytogenes infection. It was shown that orally administered viable L. casei, and not heat-killed L. casei, enhanced significantly the antigen-specific DTH at 24 and 48 h in Wistar rats, Brown Norway rats, and BALB/c mice in a time- and dose-dependent fashion. L. casei had to be administered at least 3 days prior to the DTH assay at a daily dose of 10(9) CFU in order to induce significant effects. Long-term administration of 10(9) CFU of viable L. casei resulted in enhanced ACR, as demonstrated by reduced L. monocytogenes counts in the spleen and liver and diminished serum alanine aminotransferase activity after reinfection. Enhancement of cell-mediated immunological immune responses by L. casei was further established in an adoptive transfer study. Naïve recipient BALB/c mice, which were infused with nonadherent, immunized spleen cells from L. casei-fed donor BALB/c mice, showed significantly enhanced DTH responses at 24 and 48 h compared to recipient mice which received spleen cells from control donor mice. In conclusion, orally administered L. casei enhanced cell-mediated immunological memory responses. The effects relied on lactobacillus dose and viability as well as timing of supplementation and, further, appeared to be independent of host species or genetic background.
Collapse
Affiliation(s)
- R de Waard
- Department of the Science of Food of Animal Origin, Utrecht University, Utrecht, The Netherlands.
| | | | | | | | | | | |
Collapse
|
32
|
Masihi KN, Hadden JW. Protection by methyl inosine monophosphate (MIMP) against aerosol influenza virus infection in mice. Int Immunopharmacol 2002; 2:835-41. [PMID: 12095174 DOI: 10.1016/s1567-5769(02)00026-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Methyl inosine monophosphate (MIMP) is a novel thymomimetic purine immunomodulator capable of enhancing a wide variety of immune responses. Intravenous (i.v.) administration of MIMP 1 day prior to influenza virus infection could confer partial protection and significantly increase the mean survival of treated mice. Intranasal (i.n.) administration of MIMP improved survival rates and incorporation of MIMP in squalane-saline emulsion 1 day prior to or 1 h after influenza infection conferred complete protection. Mice administered MIMP had reduced levels of lung virus and lower titers of circulating serum hemagglutination-inhibition (HI) antibodies. Complete survival and reduction of viral load after influenza challenge infection suggests effective stimulation by MIMP of protective responses against influenza virus.
Collapse
|