1
|
Borges B, Brown SM, Chen WJ, Clarke MT, Herzeg A, Park JH, Ross J, Kong L, Denton M, Smith AK, Lum T, Zada FM, Cordero M, Gupta N, Cook SE, Murray H, Matson J, Klein S, Bennett CF, Krainer AR, MacKenzie TC, Sumner CJ. Intra-amniotic antisense oligonucleotide treatment improves phenotypes in preclinical models of spinal muscular atrophy. Sci Transl Med 2025; 17:eadv4656. [PMID: 40367190 DOI: 10.1126/scitranslmed.adv4656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 04/24/2025] [Indexed: 05/16/2025]
Abstract
Neurological disorders with onset before or at birth are a leading cause of morbidity and mortality in infants and children. Prenatal treatment has the potential to reduce or prevent irreversible neuronal loss and facilitate normal neurodevelopment. We hypothesized that antisense oligonucleotides (ASOs) delivered to the amniotic fluid by intra-amniotic (IA) injection could safely distribute to the fetal central nervous system (CNS) and provide therapeutic benefit in the motor neuron disease spinal muscular atrophy (SMA), caused by mutations of the survival of motor neuron 1 gene (SMN1), leading to deficiency of SMN protein. Although the splice-switching ASO nusinersen ameliorates SMA when delivered postnatally, substantial deficits can remain in severely affected infants. Here, IA injection of ASOs into two mouse models of severe SMA increased SMN expression in the CNS. In SMAΔ7 mice, which manifest pathology in utero, prenatal treatment improved motor neuron numbers, motor axon development, motor behavioral tests, and survival when compared with those in mice treated postnatally (between P1 and P3). To assess the feasibility of prenatal treatment in a large-animal model, ASOs were delivered midgestation to fetal sheep by IA or intracranial injection. ASOs delivered by IA injection distributed to the spinal cord at therapeutic concentrations and to multiple peripheral tissues without evidence of substantial toxicity to the fetus or mother. These data demonstrated that IA delivery of ASOs holds potential as a minimally invasive approach for prenatal treatment of SMA and possibly other severe, early-onset neurological disorders.
Collapse
Affiliation(s)
- Beltran Borges
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF Center for Maternal-Fetal Precision Medicine, San Francisco, CA 94158, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Stephen M Brown
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Wan-Jin Chen
- First Affiliated Hospital of Fujian Medical University, Fuzhou 350004, China
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Maria T Clarke
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF Center for Maternal-Fetal Precision Medicine, San Francisco, CA 94158, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Akos Herzeg
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF Center for Maternal-Fetal Precision Medicine, San Francisco, CA 94158, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jae Hong Park
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Joshua Ross
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lingling Kong
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Madeline Denton
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Amy K Smith
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tony Lum
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF Center for Maternal-Fetal Precision Medicine, San Francisco, CA 94158, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Fareha Moulana Zada
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Marco Cordero
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nalin Gupta
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Pediatrics and Benioff Children's Hospital, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sarah E Cook
- Comparative Pathology Laboratory, University of California, Davis, Davis, CA 95616, USA
| | | | - John Matson
- Ionis Pharmaceuticals, Carlsbad, CA 92010, USA
| | | | | | - Adrian R Krainer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Tippi C MacKenzie
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF Center for Maternal-Fetal Precision Medicine, San Francisco, CA 94158, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Pediatrics and Benioff Children's Hospital, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Charlotte J Sumner
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
2
|
Blum KM, Turner ME, Schwarz EL, Best CA, Kelly JM, Yates AR, Hor KN, Matsuzaki Y, Drews JD, Zakko J, Shah K, Shinoka T, Humphrey JD, Marsden AL, Breuer CK. Oversized Conduits Predict Stenosis in Tissue Engineered Vascular Grafts. JACC Basic Transl Sci 2025:S2452-302X(25)00065-8. [PMID: 40243957 DOI: 10.1016/j.jacbts.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 04/18/2025]
Abstract
Tissue-engineered vascular grafts (TEVGs) offer promising advancements in treating congenital heart disease by enabling the creation of autologous tissue for complex cardiac repairs. Our approach involves implanting biodegradable scaffolds seeded with autologous cells that remodel into functional neovessels. To understand better the factors guiding neovessel formation, we evaluated 50 ovine thoracic TEVGs using angiography at 1 and 6 weeks postimplantation. Nondimensionalization accounted for anatomical differences between animals and identified hemodynamics and surgical sizing as potential driving factors. Regression analysis revealed that narrowing at the inflow anastomosis and graft oversizing correlated significantly with stenosis development. Computational fluid dynamics showed that these factors influenced wall shear stress and flow patterns, contributing to neovessel narrowing. Comparisons with clinical trial data from Fontan conduits supported these findings, emphasizing that matching graft size to the native inflow vessel can reduce stenosis and enhance TEVG performance.
Collapse
Affiliation(s)
- Kevin M Blum
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA; Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Mackenzie E Turner
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA; Department of Molecular, Cellular, and Developmental Biology, The Ohio State University, Columbus, Ohio, USA; Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Erica L Schwarz
- Department of Pediatrics and Bioengineering, Stanford University, Stanford, California, USA; Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Cameron A Best
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - John M Kelly
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA; Department of Pediatric Cardiology, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Andrew R Yates
- Department of Pediatric Cardiology, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Kan N Hor
- Department of Pediatric Cardiology, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Yuichi Matsuzaki
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Joseph D Drews
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA; Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Jason Zakko
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA; Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Kejal Shah
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA; Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Toshiharu Shinoka
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA; Department of Surgery, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Alison L Marsden
- Department of Pediatrics and Bioengineering, Stanford University, Stanford, California, USA
| | - Christopher K Breuer
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA; Department of Surgery, Nationwide Children's Hospital, Columbus, Ohio, USA.
| |
Collapse
|
3
|
Gamulin E, Mateljak Lukačević S, Lang Balija M, Smajlović A, Vnuk D, Gulan Harcet J, Tomičić M, Hećimović A, Halassy B, Kurtović T. Pharmacokinetics of Snake Antivenom Following Intravenous and Intramuscular Administration in Envenomed Large Animal Model. Pharmaceutics 2025; 17:212. [PMID: 40006579 PMCID: PMC11859798 DOI: 10.3390/pharmaceutics17020212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/27/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Background: The parenteral administration of antivenoms is the mainstay in snakebite envenoming therapy. The standardized protocol does not exist, but it is agreed that the intravenous (i.v.) route is more effective than the others, especially the intramuscular (i.m.) route, based on the monitoring of venom/antivenom pharmacokinetics in the systemic circulation. Recent evidence suggests that the lymphatic system may be crucial in abolishing venom action. Methods: A preclinical study was performed to determine the optimal administration route with emphasis on venom/antivenom interplay in both the blood and lymph of experimentally envenomed sheep. Timed level measurements were used to compare the antivenom effect on the decrement of venom quantities in both relevant body compartments. Hematological and coagulation parameters, as well as proportions of developed anti-antivenom IgGs, were evaluated. Results: The i.m. antivenom resulted in faster and greater lymphatic absorption and complete neutralization of the venom, whereas the i.v. antivenom only slowed its absorption. The total amount of venom reaching the lymph (AUC0-t) was two times lower after i.m. administration. In the systemic circulation, i.m. antivenom had a lower peak concentration (cmax) and a longer time to reach it (tmax). However, the total venom exposure was three times lower than with i.v. antivenom. Irrespective of the treatment approach, both groups showed improvement in blood disorders with no significant difference in humoral response against equine F(ab')2 fragments. Conclusions:I.m. administration proved to be a viable option for the snakebite management.
Collapse
Affiliation(s)
- Erika Gamulin
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, HR-10000 Zagreb, Croatia; (E.G.); (S.M.L.); (M.L.B.); (B.H.)
| | - Sanja Mateljak Lukačević
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, HR-10000 Zagreb, Croatia; (E.G.); (S.M.L.); (M.L.B.); (B.H.)
| | - Maja Lang Balija
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, HR-10000 Zagreb, Croatia; (E.G.); (S.M.L.); (M.L.B.); (B.H.)
| | - Ana Smajlović
- Clinic for Surgery, Orthopaedics and Ophthalmology, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, HR-10000 Zagreb, Croatia; (A.S.); (D.V.)
| | - Dražen Vnuk
- Clinic for Surgery, Orthopaedics and Ophthalmology, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, HR-10000 Zagreb, Croatia; (A.S.); (D.V.)
| | - Jadranka Gulan Harcet
- Croatian Institute of Transfusion Medicine, Petrova 3, HR-10000 Zagreb, Croatia; (J.G.H.); (M.T.); (A.H.)
| | - Maja Tomičić
- Croatian Institute of Transfusion Medicine, Petrova 3, HR-10000 Zagreb, Croatia; (J.G.H.); (M.T.); (A.H.)
| | - Ana Hećimović
- Croatian Institute of Transfusion Medicine, Petrova 3, HR-10000 Zagreb, Croatia; (J.G.H.); (M.T.); (A.H.)
| | - Beata Halassy
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, HR-10000 Zagreb, Croatia; (E.G.); (S.M.L.); (M.L.B.); (B.H.)
| | - Tihana Kurtović
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, HR-10000 Zagreb, Croatia; (E.G.); (S.M.L.); (M.L.B.); (B.H.)
| |
Collapse
|
4
|
Muniz TDTP, Rossi MC, de Vasconcelos Machado VM, Alves ALG. Mesenchymal Stem Cells and Tissue Bioengineering Applications in Sheep as Ideal Model. Stem Cells Int 2024; 2024:5176251. [PMID: 39465229 PMCID: PMC11511598 DOI: 10.1155/2024/5176251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Abstract
The most common technologies in tissue engineering include growth factor therapies; metal implants, such as titanium; 3D bioprinting; nanoimprinting for ceramic/polymer scaffolds; and cell therapies, such as mesenchymal stem cells (MSCs). Cell therapy is a promising alternative to organ grafts and transplants in the treatment of numerous musculoskeletal diseases. MSCs have increasingly been used in generative medicine due to their specialized self-renewal, immunomodulation, multiplication, and differentiation properties. To further expand the potential of these cells in tissue repair, significant efforts are currently dedicated to the production of biomaterials with desirable short- and long-term biophysical properties that can aid the differentiation and expansion of MSCs. Biomaterials support MSC differentiation by modulating their characteristics, such as composition, mechanical properties, porosity, and topography. This review aimed to describe recent MSC approaches, including those associated with biomaterials, from experimental, clinical, and preclinical studies with sheep models.
Collapse
Affiliation(s)
- Talita D'Paula Tavares Pereira Muniz
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), 18.618-681, Botucatu, Sao Paulo, Brazil
| | - Mariana Correa Rossi
- Materials Engineering Department (DEMa), São Carlos Federal University (UFSCar), 13.565-905, São Carlos, Sao Paulo, Brazil
| | - Vânia Maria de Vasconcelos Machado
- Department of Veterinary Surgery and Animal Reproduction, Imaging Diagnostic Sector, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), 18.618-681, Botucatu, Sao Paulo, Brazil
| | - Ana Liz Garcia Alves
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), 18.618-681, Botucatu, Sao Paulo, Brazil
| |
Collapse
|
5
|
Ertugrul IA, Puspitarani RADA, Wijntjes B, Vervoorn MT, Ballan EM, van der Kaaij NP, van Goor H, Westenbrink BD, van der Plaats A, Nijhuis F, van Suylen V, Erasmus ME. Ex Situ Left Ventricular Pressure-Volume Loop Analyses for Donor Hearts: Proof of Concept in an Ovine Experimental Model. Transpl Int 2024; 37:12982. [PMID: 39055346 PMCID: PMC11269103 DOI: 10.3389/ti.2024.12982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024]
Abstract
Ex situ heart perfusion (ESHP) has emerged as an important strategy to preserve donation after brain death (DBD) and donation after circulatory death (DCD) donor hearts. Clinically, both DBD and DCD hearts are successfully preserved using ESHP. Viability assessment is currently based on biochemical values, while a reliable method for graft function assessment in a physiologic working mode is unavailable. As functional assessment during ESHP has demonstrated the highest predictive value of outcome post-transplantation, this is an important area for improvement. In this study, a novel method for ex situ assessment of left ventricular function with pressure-volume loop analyses is evaluated. Ovine hearts were functionally evaluated during normothermic ESHP with the novel pressure-volume loop system. This system provides an afterload and adjustable preload to the left ventricle. By increasing the preload and measuring end-systolic elastance, the system could successfully assess the left ventricular function. End-systolic elastance at 60 min and 120 min was 2.8 ± 1.8 mmHg/mL and 2.7 ± 0.7 mmHg/mL, respectively. In this study we show a novel method for functional graft assessment with ex situ pressure-loop analyses during ESHP. When further validated, this method for pressure-volume assessments, could be used for better graft selection in both DBD and DCD donor hearts.
Collapse
Affiliation(s)
- I. A. Ertugrul
- Department of Cardiothoracic Surgery, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - R. A. D. A. Puspitarani
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | | | - M. T. Vervoorn
- Department of Cardiothoracic Surgery, University Medical Centre Utrecht, Utrecht, Netherlands
| | - E. M. Ballan
- Department of Cardiothoracic Surgery, University Medical Centre Utrecht, Utrecht, Netherlands
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
- Netherlands Heart Institute, Utrecht, Netherlands
| | - N. P. van der Kaaij
- Department of Cardiothoracic Surgery, University Medical Centre Utrecht, Utrecht, Netherlands
| | - H. van Goor
- Department of Medical Biology and Pathology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - B. D. Westenbrink
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | | | | | - V. van Suylen
- Department of Cardiothoracic Surgery, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - M. E. Erasmus
- Department of Cardiothoracic Surgery, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
6
|
Borges B, Varthaliti A, Schwab M, Clarke MT, Pivetti C, Gupta N, Cadwell CR, Guibinga G, Phillips S, Del Rio T, Ozsolak F, Imai-Leonard D, Kong L, Laird DJ, Herzeg A, Sumner CJ, MacKenzie TC. Prenatal AAV9-GFP administration in fetal lambs results in transduction of female germ cells and maternal exposure to virus. Mol Ther Methods Clin Dev 2024; 32:101263. [PMID: 38827250 PMCID: PMC11141462 DOI: 10.1016/j.omtm.2024.101263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/01/2024] [Indexed: 06/04/2024]
Abstract
Prenatal somatic cell gene therapy (PSCGT) could potentially treat severe, early-onset genetic disorders such as spinal muscular atrophy (SMA) or muscular dystrophy. Given the approval of adeno-associated virus serotype 9 (AAV9) vectors in infants with SMA by the U.S. Food and Drug Administration, we tested the safety and biodistribution of AAV9-GFP (clinical-grade and dose) in fetal lambs to understand safety and efficacy after umbilical vein or intracranial injection on embryonic day 75 (E75) . Umbilical vein injection led to widespread biodistribution of vector genomes in all examined lamb tissues and in maternal uteruses at harvest (E96 or E140; term = E150). There was robust GFP expression in brain, spinal cord, dorsal root ganglia (DRGs), without DRG toxicity and excellent transduction of diaphragm and quadriceps muscles. However, we found evidence of systemic toxicity (fetal growth restriction) and maternal exposure to the viral vector (transient elevation of total bilirubin and a trend toward elevation in anti-AAV9 antibodies). There were no antibodies against GFP in ewes or lambs. Analysis of fetal gonads demonstrated GFP expression in female (but not male) germ cells, with low levels of integration-specific reads, without integration in select proto-oncogenes. These results suggest potential therapeutic benefit of AAV9 PSCGT for neuromuscular disorders, but warrant caution for exposure of female germ cells.
Collapse
Affiliation(s)
- Beltran Borges
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF Center for Maternal-Fetal Precision Medicine, San Francisco, CA 94158, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Antonia Varthaliti
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Marisa Schwab
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Maria T Clarke
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF Center for Maternal-Fetal Precision Medicine, San Francisco, CA 94158, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Christopher Pivetti
- Department of Surgery, University of California, Davis, Davis, CA 95817, USA
| | - Nalin Gupta
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Pediatrics and Benioff Children's Hospital, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Cathryn R Cadwell
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Neurohub, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ghiabe Guibinga
- Novartis Institutes for BioMedical Research Biologics Center, San Diego, CA 92121, USA
| | - Shirley Phillips
- Novartis Institutes for BioMedical Research Biologics Center, San Diego, CA 92121, USA
| | - Tony Del Rio
- Novartis Institutes for BioMedical Research Biologics Center, San Diego, CA 92121, USA
| | - Fatih Ozsolak
- Novartis Institutes for BioMedical Research Biologics Center, San Diego, CA 92121, USA
| | - Denise Imai-Leonard
- Comparative Pathology Laboratory, University of California, Davis, Davis, CA 95616, USA
| | - Lingling Kong
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Diana J Laird
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Obstetrics and Gynecology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Akos Herzeg
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF Center for Maternal-Fetal Precision Medicine, San Francisco, CA 94158, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Charlotte J Sumner
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Tippi C MacKenzie
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF Center for Maternal-Fetal Precision Medicine, San Francisco, CA 94158, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Pediatrics and Benioff Children's Hospital, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Obstetrics and Gynecology, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
7
|
Yan Z, Yang J, Wei WT, Zhou ML, Mo DX, Wan X, Ma R, Wu MM, Huang JH, Liu YJ, Lv FH, Li MH. A time-resolved multi-omics atlas of transcriptional regulation in response to high-altitude hypoxia across whole-body tissues. Nat Commun 2024; 15:3970. [PMID: 38730227 PMCID: PMC11087590 DOI: 10.1038/s41467-024-48261-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 04/23/2024] [Indexed: 05/12/2024] Open
Abstract
High-altitude hypoxia acclimatization requires whole-body physiological regulation in highland immigrants, but the underlying genetic mechanism has not been clarified. Here we use sheep as an animal model for low-to-high altitude translocation. We generate multi-omics data including whole-genome sequences, time-resolved bulk RNA-Seq, ATAC-Seq and single-cell RNA-Seq from multiple tissues as well as phenotypic data from 20 bio-indicators. We characterize transcriptional changes of all genes in each tissue, and examine multi-tissue temporal dynamics and transcriptional interactions among genes. Particularly, we identify critical functional genes regulating the short response to hypoxia in each tissue (e.g., PARG in the cerebellum and HMOX1 in the colon). We further identify TAD-constrained cis-regulatory elements, which suppress the transcriptional activity of most genes under hypoxia. Phenotypic and transcriptional evidence indicate that antenatal hypoxia could improve hypoxia tolerance in offspring. Furthermore, we provide time-series expression data of candidate genes associated with human mountain sickness (e.g., BMPR2) and high-altitude adaptation (e.g., HIF1A). Our study provides valuable resources and insights for future hypoxia-related studies in mammals.
Collapse
Affiliation(s)
- Ze Yan
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ji Yang
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Wen-Tian Wei
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ming-Liang Zhou
- Sichuan Academy of Grassland Science, Chengdu, 611743, China
| | - Dong-Xin Mo
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xing Wan
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Rui Ma
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Mei-Ming Wu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jia-Hui Huang
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ya-Jing Liu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Feng-Hua Lv
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Meng-Hua Li
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China.
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
8
|
Shah AA, Sheikh AA, Hasin D, Shah F, Aarif O, Shah RA, Ahmad SB, Maqbool S, Pampori ZA. Isolation, in vitro expansion and characterization of ovine fetal adnexa-derived mesenchymal stem cells reveals a source dependent trilineage differentiation and growth kinetics. Anim Biotechnol 2023; 34:3908-3919. [PMID: 37493347 DOI: 10.1080/10495398.2023.2238015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
This study was designed to isolate, cultivate, characterize and evaluate the growth kinetics of mesenchymal stem cells (MSCs) derived from fetal adnexa of sheep. The gravid uteri of ewes were collected from a local abattoir. The MSCs isolated from different fetal regions (Wharton's Jelly [oWJ], cord blood [oCB], amniotic fluid [oAF] and amniotic Sac [oAS]) were expanded in vitro and characterized for surface and pluripotency markers. The growth kinetics of MSCs was compared at 3rd and 5th passages. Similarly, the colony-forming efficiency (CFE) assay was performed at 3rd passage. The fetal adnexa-derived ovine MSCs showed the expression of CD73, CD90 and CD105. Similarly, the MSCs also expressed pluripotency markers, OCT4 and SOX2. Besides, cells also differentiated into osteogenic, chondrogenic and adipogenic lineages. The MSCs in culture showed a typical growth curve with initial lag phase, an exponential phase, a plateau phase and a decline phase. The growth rate was highest in oAF-MSCs at P5. The population doubling time (PDT) was highest in oAS-MSCs (87.28 ± 3.24 h), whereas the colony number was highest in oAF-MSCs (53.67 ± 4.06). The study reveals that oAF-MSCs were superior which outperformed other MSCs indicating that oAF-derived MSCs could be utilized for regenerative medicine.
Collapse
Affiliation(s)
- Aamir Amin Shah
- Division of Veterinary Physiology, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-Kashmir, Shuhama, J & K, India
| | - Aasif Ahmad Sheikh
- Division of Veterinary Physiology, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-Kashmir, Shuhama, J & K, India
| | - Dilruba Hasin
- Division of Veterinary Physiology, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-Kashmir, Shuhama, J & K, India
| | - Fozia Shah
- Division of Veterinary Physiology, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-Kashmir, Shuhama, J & K, India
| | - Ovais Aarif
- Division of Veterinary Physiology, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-Kashmir, Shuhama, J & K, India
| | - Riaz Ahmad Shah
- Division of Animal Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-Kashmir, Shuhama, J & K, India
| | - Sheikh Bilal Ahmad
- Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-Kashmir, Shuhama, J & K, India
| | - Showkat Maqbool
- Division of Animal Genetics and Breeding, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-Kashmir, Shuhama, J & K, India
| | - Z A Pampori
- Division of Veterinary Physiology, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-Kashmir, Shuhama, J & K, India
| |
Collapse
|
9
|
Murray SJ, Wellby MP, Barrell GK, Russell KN, Deane AR, Wynyard JR, Gray SJ, Palmer DN, Mitchell NL. Efficacy of dual intracerebroventricular and intravitreal CLN5 gene therapy in sheep prompts the first clinical trial to treat CLN5 Batten disease. Front Pharmacol 2023; 14:1212235. [PMID: 37942487 PMCID: PMC10628725 DOI: 10.3389/fphar.2023.1212235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/29/2023] [Indexed: 11/10/2023] Open
Abstract
Mutations in the CLN5 gene cause the fatal, pediatric, neurodegenerative disease CLN5 neuronal ceroid lipofuscinosis. Affected children suffer progressive neuronal loss, visual failure and premature death. Presently there is no treatment. This study evaluated dual intracerebroventricular (ICV) and intravitreal (IVT) administration of a self-complementary adeno-associated viral vector encoding ovine CLN5 (scAAV9/oCLN5) into CLN5 affected sheep (CLN5-/-) at various disease stages. CLN5 disease progression was slowed in pre-symptomatic sheep who received a moderate dose of scAAV9/oCLN5, whilst a higher ICV dose treatment in early and advanced symptomatic animals delayed or halted disease progression. Intracranial (brain) volume loss was attenuated in all treatment cohorts, and visual function was also sustained in both the early and advanced symptomatic treated sheep over the 24-month duration of the study. Robust CLN5 protein expression was detected throughout the brain and spinal cord, and improvements in central nervous system and retinal disease correlates were observed. These findings hold translational promise for extending and improving the quality of life in both pre-symptomatic and symptomatic CLN5 patients, and prompted the initiation of the first in-human Phase I/II clinical trial testing ICV/IVT administration of scAAV9 encoding human CLN5 (https://clinicaltrials.gov/; NCT05228145).
Collapse
Affiliation(s)
- Samantha J. Murray
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Martin P. Wellby
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Graham K. Barrell
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Katharina N. Russell
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Ashley R. Deane
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - John R. Wynyard
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| | - Steven J. Gray
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - David N. Palmer
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
- Department of Radiology, University of Otago, Christchurch, New Zealand
| | - Nadia L. Mitchell
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
- Department of Radiology, University of Otago, Christchurch, New Zealand
| |
Collapse
|
10
|
Miao Y, Zhao X, Lei J, Ding J, Feng H, Wu K, Liu J, Wang C, Ye D, Wang X, Wang J, Yang Z. Characterization of Lung Microbiomes in Pneumonic Hu Sheep Using Culture Technique and 16S rRNA Gene Sequencing. Animals (Basel) 2023; 13:2763. [PMID: 37685027 PMCID: PMC10486422 DOI: 10.3390/ani13172763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/13/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Hu sheep, a locally bred species in China known for its high productivity, is currently suffering from pneumonia. Here, we combine high-throughput 16SrRNA gene sequencing and bacterial culturing to examine the bacterial community in pneumonic Hu Sheep lungs (p < 0.05). The results showed that the abundance and diversity of lung bacteria in healthy sheep were significantly higher than those in pneumonia sheep (p = 0.139), while there was no significant difference between moderate and severe pneumonia. Furthermore, the composition of the lung microbiota community underwent significant alterations between different levels of pneumonia severity. The application of LEfSe analysis revealed a notable enrichment of Mannheimiae within the lungs of sheep afflicted with moderate pneumonia (p < 0.01), surpassing the levels observed in their healthy counterparts. Additionally, Fusobacterium emerged as the prevailing bacterial group within the lungs of sheep suffering from severe pneumonia. Integrating the results of bacterial isolation and identification, we conclusively determined that Mannheimia haemolytica was the primary pathogenic bacterium within the lungs of sheep afflicted with moderate pneumonia. Furthermore, the exacerbation of pneumonia may be attributed to the synergistic interplay between Fusobacterium spp. and other bacterial species. Our results provide new insights for guiding preventive and therapeutic measures for pneumonia of different severities in sheep.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Zengqi Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (Y.M.); (X.Z.); (J.L.); (J.D.); (H.F.); (K.W.); (C.W.); (X.W.); (J.W.)
| |
Collapse
|
11
|
Woodrow JS, Sheats MK, Cooper B, Bayless R. Asthma: The Use of Animal Models and Their Translational Utility. Cells 2023; 12:cells12071091. [PMID: 37048164 PMCID: PMC10093022 DOI: 10.3390/cells12071091] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/14/2023] Open
Abstract
Asthma is characterized by chronic lower airway inflammation that results in airway remodeling, which can lead to a permanent decrease in lung function. The pathophysiology driving the development of asthma is complex and heterogenous. Animal models have been and continue to be essential for the discovery of molecular pathways driving the pathophysiology of asthma and novel therapeutic approaches. Animal models of asthma may be induced or naturally occurring. Species used to study asthma include mouse, rat, guinea pig, cat, dog, sheep, horse, and nonhuman primate. Some of the aspects to consider when evaluating any of these asthma models are cost, labor, reagent availability, regulatory burden, relevance to natural disease in humans, type of lower airway inflammation, biological samples available for testing, and ultimately whether the model can answer the research question(s). This review aims to discuss the animal models most available for asthma investigation, with an emphasis on describing the inciting antigen/allergen, inflammatory response induced, and its translation to human asthma.
Collapse
Affiliation(s)
- Jane Seymour Woodrow
- Department of Clinical Studies, New Bolton Center, College of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA 19348, USA
| | - M Katie Sheats
- Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27606, USA
| | - Bethanie Cooper
- Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27606, USA
| | - Rosemary Bayless
- Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27606, USA
| |
Collapse
|
12
|
Li Y, Lian D, Wang J, Zhao Y, Li Y, Liu G, Wu S, Deng S, Du X, Lian Z. MDM2 antagonists promote CRISPR/Cas9-mediated precise genome editing in sheep primary cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:309-323. [PMID: 36726409 PMCID: PMC9883270 DOI: 10.1016/j.omtn.2022.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/31/2022] [Indexed: 01/04/2023]
Abstract
CRISPR-Cas9-mediated genome editing in sheep is of great use in both agricultural and biomedical applications. While targeted gene knockout by CRISPR-Cas9 through non-homologous end joining (NHEJ) has worked efficiently, the knockin efficiency via homology-directed repair (HDR) remains lower, which severely hampers the application of precise genome editing in sheep. Here, in sheep fetal fibroblasts (SFFs), we optimized several key parameters that affect HDR, including homology arm (HA) length and the amount of double-stranded DNA (dsDNA) repair template; we also observed synchronization of SFFs in G2/M phase could increase HDR efficiency. Besides, we identified three potent small molecules, RITA, Nutlin3, and CTX1, inhibitors of p53-MDM2 interaction, that caused activation of the p53 pathway, resulting in distinct G2/M cell-cycle arrest in response to DNA damage and improved CRISPR-Cas9-mediated HDR efficiency by 1.43- to 4.28-fold in SFFs. Furthermore, we demonstrated that genetic knockout of p53 could inhibit HDR in SFFs by suppressing the expression of several key factors involved in the HDR pathway, such as BRCA1 and RAD51. Overall, this study offers an optimized strategy for the usage of dsDNA repair template, more importantly, the application of MDM2 antagonists provides a simple and efficient strategy to promote CRISPR/Cas9-mediated precise genome editing in sheep primary cells.
Collapse
Affiliation(s)
- Yan Li
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China,State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China,Laboratory Animal Center of the Academy of Military Medical Sciences, Beijing 100071, China,These authors contributed equally
| | - Di Lian
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China,These authors contributed equally
| | - Jiahao Wang
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China,Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,These authors contributed equally
| | - Yue Zhao
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yao Li
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Guoshi Liu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Sen Wu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Shoulong Deng
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China,Corresponding author: Shoulong Deng, NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, 5 Panjiayuannanli, Chaoyang District, Beijing 100021, China.
| | - Xuguang Du
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China,Corresponding author: Xuguang Du, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Zhengxing Lian
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China,Corresponding author: Zhengxing Lian, Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, 2 Mingyuanxilu, Haidian District, Beijing 100193, China. .
| |
Collapse
|
13
|
Wu H, Zhou W, Liu H, Cui X, Ma W, Wu H, Li G, Wang L, Zhang J, Zhang X, Ji P, Lian Z, Liu G. Whole-genome methylation analysis reveals epigenetic variation between wild-type and nontransgenic cloned, ASMT transgenic cloned dairy goats generated by the somatic cell nuclear transfer. J Anim Sci Biotechnol 2022; 13:145. [PMID: 36434676 PMCID: PMC9701027 DOI: 10.1186/s40104-022-00764-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 08/03/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND SCNT (somatic cell nuclear transfer) is of great significance to biological research and also to the livestock breeding. However, the survival rate of the SCNT cloned animals is relatively low compared to other transgenic methods. This indicates the potential epigenetic variations between them. DNA methylation is a key marker of mammalian epigenetics and its alterations will lead to phenotypic differences. In this study, ASMT (acetylserotonin-O-methyltransferase) ovarian overexpression transgenic goat was produced by using SCNT. To investigate whether there are epigenetic differences between cloned and WT (wild type) goats, WGBS (whole-genome bisulfite sequencing) was used to measure the whole-genome methylation of these animals. RESULTS It is observed that the different mCpG sites are mainly present in the intergenic and intronic regions between cloned and WT animals, and their CG-type methylation sites are strongly correlated. DMR (differentially methylated region) lengths are located around 1000 bp, mainly distributed in the exonic, intergenic and intronic functional domains. A total of 56 and 36 DMGs (differentially methylated genes) were identified by GO and KEGG databases, respectively. Functional annotation showed that DMGs were enriched in biological-process, cellular-component, molecular-function and other signaling pathways. A total of 10 identical genes related to growth and development were identified in GO and KEGG databases. CONCLUSION The differences in methylation genes among the tested animals have been identified. A total of 10 DMGs associated with growth and development were identified between cloned and WT animals. The results indicate that the differential patterns of DNA methylation between the cloned and WT goats are probably caused by the SCNT. These novel observations will help us to further identify the unveiled mechanisms of somatic cell cloning technology, particularly in goats.
Collapse
Affiliation(s)
- Hao Wu
- grid.22935.3f0000 0004 0530 8290National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China ,Sany Institute of China Agricultural University, Sanya, 572025 China
| | - Wendi Zhou
- grid.22935.3f0000 0004 0530 8290National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Haijun Liu
- Institute of Animal Husbandry and Veterinary, Academy of Agricultural Sciences of Tianjin, Tianjin, 300192 China
| | - Xudai Cui
- Qingdao Senmiao Industrial Co., Ltd., Qingdao, 266101 China
| | - Wenkui Ma
- grid.22935.3f0000 0004 0530 8290National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Haixin Wu
- grid.22935.3f0000 0004 0530 8290National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Guangdong Li
- grid.22935.3f0000 0004 0530 8290National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Likai Wang
- grid.22935.3f0000 0004 0530 8290National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Jinlong Zhang
- Institute of Animal Husbandry and Veterinary, Academy of Agricultural Sciences of Tianjin, Tianjin, 300192 China
| | - Xiaosheng Zhang
- Institute of Animal Husbandry and Veterinary, Academy of Agricultural Sciences of Tianjin, Tianjin, 300192 China
| | - Pengyun Ji
- grid.22935.3f0000 0004 0530 8290National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Zhengxing Lian
- grid.22935.3f0000 0004 0530 8290National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Guoshi Liu
- grid.22935.3f0000 0004 0530 8290National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China ,Sany Institute of China Agricultural University, Sanya, 572025 China
| |
Collapse
|
14
|
Cai L, Xu H, Cui Z. Factors Limiting the Translatability of Rodent Model-Based Intranasal Vaccine Research to Humans. AAPS PharmSciTech 2022; 23:191. [PMID: 35819736 PMCID: PMC9274968 DOI: 10.1208/s12249-022-02330-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 06/09/2022] [Indexed: 12/19/2022] Open
Abstract
The intranasal route of vaccination presents an attractive alternative to parenteral routes and offers numerous advantages, such as the induction of both mucosal and systemic immunity, needle-free delivery, and increased patient compliance. Despite demonstrating promising results in preclinical studies, however, few intranasal vaccine candidates progress beyond early clinical trials. This discrepancy likely stems in part from the limited predictive value of rodent models, which are used frequently in intranasal vaccine research. In this review, we explored the factors that limit the translatability of rodent-based intranasal vaccine research to humans, focusing on the differences in anatomy, immunology, and disease pathology between rodents and humans. We also discussed approaches that minimize these differences and examined alternative animal models that would produce more clinically relevant research.
Collapse
Affiliation(s)
- Lucy Cai
- University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, USA
| | - Haiyue Xu
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, 2409 University Ave., A1900, Austin, Texas, 78712, USA
| | - Zhengrong Cui
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, 2409 University Ave., A1900, Austin, Texas, 78712, USA.
| |
Collapse
|
15
|
Burmeister DM, Supp DM, Clark RA, Tredget EE, Powell HM, Enkhbaatar P, Bohannon JK, Cancio LC, Hill DM, Nygaard RM. Advantages and Disadvantages of Using Small and Large Animals in Burn Research: Proceedings of the 2021 Research Special Interest Group. J Burn Care Res 2022; 43:1032-1041. [PMID: 35778269 DOI: 10.1093/jbcr/irac091] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Multiple animal species and approaches have been used for modeling different aspects of burn care, with some strategies considered more appropriate or translatable than others. On April 15, 2021, the Research Special Interest Group of the American Burn Association held a virtual session as part of the agenda for the annual meeting. The session was set up as a pro/con debate on the use of small versus large animals for application to four important aspects of burn pathophysiology: burn healing/conversion; scarring; inhalation injury; and sepsis. For each of these topics, 2 experienced investigators (one each for small and large animal models) described the advantages and disadvantages of using these preclinical models. The use of swine as a large animal model was a common theme due to anatomic similarities with human skin. The exception to this was a well-defined ovine model of inhalation injury; both of these species have larger airways which allow for incorporation of clinical tools such as bronchoscopes. However, these models are expensive and demanding from labor and resource standpoints. Various strategies have been implemented to make the more inexpensive rodent models appropriate for answering specific questions of interest in burns. Moreover, modelling burn-sepsis in large animals has proven difficult. It was agreed that the use of both small and large animal models have merit for answering basic questions about the responses to burn injury. Expert opinion and the ensuing lively conversations are summarized herein, which we hope will help inform experimental design of future research.
Collapse
Affiliation(s)
- David M Burmeister
- Uniformed Services University of the Health Sciences, Department of Medicine, Bethesda, MD, United States of America
| | - Dorothy M Supp
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Scientific Staff, Shriners Children's Ohio, Dayton, OH, USA
| | - Richard A Clark
- Stony Brook University, Departments of Dermatology, Biomedical Engineering and Medicine, Stony Brook, NY, USA
| | - Edward E Tredget
- Firefighters' Burn Treatment Unit, Department of Surgery, 2D3.31 Mackenzie Health Sciences Centre, University of Alberta, Edmonton, AB, Canada
| | - Heather M Powell
- Department of Materials Science and Engineering, Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA.,Scientific Staff, Shriners Children's Ohio, Dayton, OH, USA
| | - Perenlei Enkhbaatar
- Department of Anesthesiology, Medical Branch, University of Texas, 301 University Boulevard, Galveston, TX, USA
| | - Julia K Bohannon
- Vanderbilt University Medical Center, Department of Anesthesiology, Department of Pathology, Microbiology, and Immunology, Nashville, TN, USA
| | - Leopoldo C Cancio
- United States Army Institute of Surgical Research, JBSA Fort Sam Houston, TX, USA
| | - David M Hill
- Firefighters' Burn Center, Regional One Health, 877 Jefferson Avenue, Memphis, TN, USA
| | - Rachel M Nygaard
- Department of Surgery, Hennepin Healthcare, Minneapolis, MN, USA
| |
Collapse
|
16
|
Stevenson TJ, Liddle TA, Stewart C, Marshall CJ, Majumdar G. Neural programming of seasonal physiology in birds and mammals: A modular perspective. Horm Behav 2022; 142:105153. [PMID: 35325691 DOI: 10.1016/j.yhbeh.2022.105153] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/30/2022] [Accepted: 03/07/2022] [Indexed: 11/17/2022]
Abstract
Most animals in the temperate zone exhibit robust seasonal rhythms in neuroendocrine, physiological and behavioral processes. The integration of predictive and supplementary environmental cues (e.g., nutrients) involves a series of discrete, and interconnected brain regions that span hypothalamic, thalamic, mesencephalic, and limbic regions. Species-specific adaptive changes in these neuroendocrine structures and cellular plasticity have likely evolved to support seasonal life-history transitions. Despite significant advances in our understanding of ecological responses to predictive and supplementary environmental cues, there remains a paucity of literature on how these diverse cues impact the underlying neural and cellular substrates. To date, most scientific approach has focused on neuroendocrine responses to annual changes in daylength, referred to as photoperiod, due to the robust physiological changes to light manipulations in laboratory settings. In this review, we highlight the relatively few animal models that have been effectively used to investigate how predictive day lengths, and supplementary cues are integrated across hypothalamic nuclei, and discuss key findings of how seasonal rhythms in physiology are governed by adaptive neuroendocrine changes. We discuss how specific brain regions integrate environmental cues to form a complex multiunit or 'modular' system that has evolved to optimize the timing of seasonal physiology. Overall, the review aims to highlight the existence of a modular network of neural regions that independently contribute to timing seasonal physiology. This paper proposes that a multi-modular neuroendocrine system has evolved in which independent neural 'units' operate to support species-specific seasonal rhythms.
Collapse
Affiliation(s)
- Tyler J Stevenson
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G61 1QH, United Kingdom.
| | - Timothy A Liddle
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G61 1QH, United Kingdom
| | - Calum Stewart
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G61 1QH, United Kingdom
| | - Christopher J Marshall
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G61 1QH, United Kingdom
| | - Gaurav Majumdar
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G61 1QH, United Kingdom
| |
Collapse
|
17
|
Abstract
For four decades, genetically altered laboratory animals have provided invaluable information. Originally, genetic modifications were performed on only a few animal species, often chosen because of the ready accessibility of embryonic materials and short generation times. The methods were often slow, inefficient and expensive. In 2013, a new, extremely efficient technology, namely CRISPR/Cas9, not only made the production of genetically altered organisms faster and cheaper, but also opened it up to non-conventional laboratory animal species. CRISPR/Cas9 relies on a guide RNA as a 'location finder' to target DNA double strand breaks induced by the Cas9 enzyme. This is a prerequisite for non-homologous end joining repair to occur, an error prone mechanism often generating insertion or deletion of genetic material. If a DNA template is also provided, this can lead to homology directed repair, allowing precise insertions, deletions or substitutions. Due to its high efficiency in targeting DNA, CRISPR/Cas9-mediated genetic modification is now possible in virtually all animal species for which we have genome sequence data. Furthermore, modifications of Cas9 have led to more refined genetic alterations from targeted single base-pair mutations to epigenetic modifications. The latter offer altered gene expression without genome alteration. With this ever growing genetic toolbox, the number and range of genetically altered conventional and non-conventional laboratory animals with simple or complex genetic modifications is growing exponentially.
Collapse
|
18
|
See Hoe LE, Wildi K, Obonyo NG, Bartnikowski N, McDonald C, Sato K, Heinsar S, Engkilde-Pedersen S, Diab S, Passmore MR, Wells MA, Boon AC, Esguerra A, Platts DG, James L, Bouquet M, Hyslop K, Shuker T, Ainola C, Colombo SM, Wilson ES, Millar JE, Malfertheiner MV, Reid JD, O'Neill H, Livingstone S, Abbate G, Sato N, He T, von Bahr V, Rozencwajg S, Byrne L, Pimenta LP, Marshall L, Nair L, Tung JP, Chan J, Haqqani H, Molenaar P, Li Bassi G, Suen JY, McGiffin DC, Fraser JF. A clinically relevant sheep model of orthotopic heart transplantation 24 h after donor brainstem death. Intensive Care Med Exp 2021; 9:60. [PMID: 34950993 PMCID: PMC8702587 DOI: 10.1186/s40635-021-00425-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/23/2021] [Indexed: 11/10/2022] Open
Abstract
Background Heart transplantation (HTx) from brainstem dead (BSD) donors is the gold-standard therapy for severe/end-stage cardiac disease, but is limited by a global donor heart shortage. Consequently, innovative solutions to increase donor heart availability and utilisation are rapidly expanding. Clinically relevant preclinical models are essential for evaluating interventions for human translation, yet few exist that accurately mimic all key HTx components, incorporating injuries beginning in the donor, through to the recipient. To enable future assessment of novel perfusion technologies in our research program, we thus aimed to develop a clinically relevant sheep model of HTx following 24 h of donor BSD.
Methods BSD donors (vs. sham neurological injury, 4/group) were hemodynamically supported and monitored for 24 h, followed by heart preservation with cold static storage. Bicaval orthotopic HTx was performed in matched recipients, who were weaned from cardiopulmonary bypass (CPB), and monitored for 6 h. Donor and recipient blood were assayed for inflammatory and cardiac injury markers, and cardiac function was assessed using echocardiography. Repeated measurements between the two different groups during the study observation period were assessed by mixed ANOVA for repeated measures.
Results Brainstem death caused an immediate catecholaminergic hemodynamic response (mean arterial pressure, p = 0.09), systemic inflammation (IL-6 - p = 0.025, IL-8 - p = 0.002) and cardiac injury (cardiac troponin I, p = 0.048), requiring vasopressor support (vasopressor dependency index, VDI, p = 0.023), with normalisation of biomarkers and physiology over 24 h. All hearts were weaned from CPB and monitored for 6 h post-HTx, except one (sham) recipient that died 2 h post-HTx. Hemodynamic (VDI - p = 0.592, heart rate - p = 0.747) and metabolic (blood lactate, p = 0.546) parameters post-HTx were comparable between groups, despite the observed physiological perturbations that occurred during donor BSD. All p values denote interaction among groups and time in the ANOVA for repeated measures. Conclusions We have successfully developed an ovine HTx model following 24 h of donor BSD. After 6 h of critical care management post-HTx, there were no differences between groups, despite evident hemodynamic perturbations, systemic inflammation, and cardiac injury observed during donor BSD. This preclinical model provides a platform for critical assessment of injury development pre- and post-HTx, and novel therapeutic evaluation. Supplementary Information The online version contains supplementary material available at 10.1186/s40635-021-00425-4.
Collapse
Affiliation(s)
- Louise E See Hoe
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia. .,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia. .,School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia.
| | - Karin Wildi
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,Cardiovascular Research Institute Basel, Basel, Switzerland
| | - Nchafatso G Obonyo
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,Wellcome Trust Centre for Global Health Research, Imperial College London, London, UK.,Initiative to Develop African Research Leaders (IDeAL), Kilifi, Kenya
| | - Nicole Bartnikowski
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Charles McDonald
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Department of Anaesthesia and Perfusion, The Prince Charles Hospital, Chermside, QLD, Australia
| | - Kei Sato
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Silver Heinsar
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,Second Department of Intensive Care, North Estonia Medical Centre, Tallinn, Estonia
| | - Sanne Engkilde-Pedersen
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Research and Development, Australian Red Cross Lifeblood, Brisbane, QLD, Australia
| | - Sara Diab
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Margaret R Passmore
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Matthew A Wells
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
| | - Ai-Ching Boon
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Arlanna Esguerra
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Research and Development, Australian Red Cross Lifeblood, Brisbane, QLD, Australia
| | - David G Platts
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Lynnette James
- Department of Cardiac Surgery, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Mahe Bouquet
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Kieran Hyslop
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Tristan Shuker
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Carmen Ainola
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Sebastiano M Colombo
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,Department of Pathophysiology and Transplantation, Università Degli Studi di Milano, Milan, Italy
| | - Emily S Wilson
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Jonathan E Millar
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Maximillian V Malfertheiner
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Department of Internal Medicine II, Cardiology and Pneumology, University Medical Center Regensburg, Regensburg, Germany
| | - Janice D Reid
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Hollier O'Neill
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Samantha Livingstone
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Gabriella Abbate
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Noriko Sato
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Ting He
- Department of Cardiac Surgery, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Viktor von Bahr
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sacha Rozencwajg
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Pitié-Salpêtrière University Hospital, Paris, France
| | - Liam Byrne
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,The Canberra Hospital Intensive Care, Garran, ACT, Australia.,Australia National University, Canberra, ACT, Australia
| | - Leticia P Pimenta
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Lachlan Marshall
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Department of Cardiac Surgery, Princess Alexandra Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital, Brisbane, QLD, Australia
| | - Lawrie Nair
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital, Brisbane, QLD, Australia
| | - John-Paul Tung
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,Research and Development, Australian Red Cross Lifeblood, Brisbane, QLD, Australia.,Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Jonathan Chan
- Prince Charles Hospital, Brisbane, QLD, Australia.,School of Medicine, Griffith University, Southport, QLD, Australia
| | - Haris Haqqani
- Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,Prince Charles Hospital, Brisbane, QLD, Australia
| | - Peter Molenaar
- Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Gianluigi Li Bassi
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain
| | - Jacky Y Suen
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - David C McGiffin
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Cardiothoracic Surgery and Transplantation, The Alfred Hospital, Melbourne, VIC, Australia.,Monash University, Melbourne, VIC, Australia
| | - John F Fraser
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
19
|
Karrow NA, Shandilya UK, Pelech S, Wagter-Lesperance L, McLeod D, Bridle B, Mallard BA. Maternal COVID-19 Vaccination and Its Potential Impact on Fetal and Neonatal Development. Vaccines (Basel) 2021; 9:1351. [PMID: 34835282 PMCID: PMC8617890 DOI: 10.3390/vaccines9111351] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/09/2021] [Accepted: 11/15/2021] [Indexed: 12/17/2022] Open
Abstract
Vaccines have been developed at "warp speed" to combat the COVID-19 pandemic caused by the SARS-CoV-2 coronavirus. Although they are considered the best approach for preventing mortality, when assessing the safety of these vaccines, pregnant women have not been included in clinical trials. Thus, vaccine safety for this demographic, as well as for the developing fetus and neonate, remains to be determined. A global effort has been underway to encourage pregnant women to get vaccinated despite the uncertain risk posed to them and their offspring. Given this, post-hoc data collection, potentially for years, will be required to determine the outcomes of COVID-19 and vaccination on the next generation. Most COVID-19 vaccine reactions include injection site erythema, pain, swelling, fatigue, headache, fever and lymphadenopathy, which may be sufficient to affect fetal/neonatal development. In this review, we have explored components of the first-generation viral vector and mRNA COVID-19 vaccines that are believed to contribute to adverse reactions and which may negatively impact fetal and neonatal development. We have followed this with a discussion of the potential for using an ovine model to explore the long-term outcomes of COVID-19 vaccination during the prenatal and neonatal periods.
Collapse
Affiliation(s)
- Niel A. Karrow
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Umesh K. Shandilya
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Steven Pelech
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada;
| | - Lauraine Wagter-Lesperance
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.W.-L.); (B.B.); (B.A.M.)
| | - Deanna McLeod
- Kaleidoscope Strategic Inc., Toronto, ON M6R 1E7, Canada;
| | - Byram Bridle
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.W.-L.); (B.B.); (B.A.M.)
| | - Bonnie A. Mallard
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.W.-L.); (B.B.); (B.A.M.)
| |
Collapse
|
20
|
Varma R, Marin‐Araujo AE, Rostami S, Waddell TK, Karoubi G, Haykal S. Short-Term Preclinical Application of Functional Human Induced Pluripotent Stem Cell-Derived Airway Epithelial Patches. Adv Healthc Mater 2021; 10:e2100957. [PMID: 34569180 DOI: 10.1002/adhm.202100957] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/15/2021] [Indexed: 12/17/2022]
Abstract
Airway pathologies including cancer, trauma, and stenosis lack effective treatments, meanwhile airway transplantation and available tissue engineering approaches fail due to epithelial dysfunction. Autologous progenitors do not meet the clinical need for regeneration due to their insufficient expansion and differentiation, for which human induced pluripotent stem cells (hiPSCs) are promising alternatives. Airway epithelial patches are engineered by differentiating hiPSC-derived airway progenitors into physiological proportions of ciliated (73.9 ± 5.5%) and goblet (2.1 ± 1.4%) cells on a silk fibroin-collagen vitrigel membrane (SF-CVM) composite biomaterial for transplantation in porcine tracheal defects ex vivo and in vivo. Evaluation of ex vivo tracheal repair using hiPSC-derived SF-CVM patches demonstrate native-like tracheal epithelial metabolism and maintenance of mucociliary epithelium to day 3. In vivo studies demonstrate SF-CVM integration and maintenance of airway patency, showing 80.8 ± 3.6% graft coverage with an hiPSC-derived pseudostratified epithelium and 70.7 ± 2.3% coverage with viable cells, 3 days postoperatively. The utility of bioengineered, hiPSC-derived epithelial patches for airway repair is demonstrated in a short-term preclinical survival model, providing a significant leap for airway reconstruction approaches.
Collapse
Affiliation(s)
- Ratna Varma
- Latner Thoracic Surgery Laboratories Toronto General Hospital Research Institute University Health Network Toronto General Hospital University of Toronto 101 College St Toronto ON M5G 0A3 Canada
- Institute of Biomedical Engineering (BME) University of Toronto 164 College St Toronto ON M5S 3G9 Canada
| | - Alba E. Marin‐Araujo
- Latner Thoracic Surgery Laboratories Toronto General Hospital Research Institute University Health Network Toronto General Hospital University of Toronto 101 College St Toronto ON M5G 0A3 Canada
| | - Sara Rostami
- Latner Thoracic Surgery Laboratories Toronto General Hospital Research Institute University Health Network Toronto General Hospital University of Toronto 101 College St Toronto ON M5G 0A3 Canada
| | - Thomas K. Waddell
- Latner Thoracic Surgery Laboratories Toronto General Hospital Research Institute University Health Network Toronto General Hospital University of Toronto 101 College St Toronto ON M5G 0A3 Canada
- Institute of Biomedical Engineering (BME) University of Toronto 164 College St Toronto ON M5S 3G9 Canada
- Institute of Medical Sciences University of Toronto 27 King's College Cir Toronto ON M5S 1A8 Canada
| | - Golnaz Karoubi
- Latner Thoracic Surgery Laboratories Toronto General Hospital Research Institute University Health Network Toronto General Hospital University of Toronto 101 College St Toronto ON M5G 0A3 Canada
- Department of Mechanical and Industrial Engineering University of Toronto 5 King's College Circle Toronto ON M5S 3G8 Canada
- Department of Laboratory Medicine and Pathobiology University of Toronto 1 King's College Circle Toronto ON M5S 1A8 Canada
| | - Siba Haykal
- Latner Thoracic Surgery Laboratories Toronto General Hospital Research Institute University Health Network Toronto General Hospital University of Toronto 101 College St Toronto ON M5G 0A3 Canada
- Institute of Medical Sciences University of Toronto 27 King's College Cir Toronto ON M5S 1A8 Canada
- Division of Plastic and Reconstructive Surgery Department of Surgery University of Toronto 200 Elizabeth Street 8N‐869 Toronto ON M5G2P7 Canada
| |
Collapse
|
21
|
Šturm S, Švara T, Spörndly-Nees E, Cerkvenik-Flajs V, Gombač M, Weber AL, Weber K. Seminiferous epithelium cycle staging based on the development of the acrosome in ram testis. J Toxicol Pathol 2021; 34:331-338. [PMID: 34621108 PMCID: PMC8484928 DOI: 10.1293/tox.2021-0025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/29/2021] [Indexed: 11/19/2022] Open
Abstract
Testicular histopathology is considered the most sensitive and reliable method to detect the effects of chemicals on sperm production. To carry out a sensitive examination of testicular histopathology and interpret the changes require knowledge of spermatogenic stages. Spermatogenic staging based on acrosome development during spermiogenesis is conventionally performed in animal species routinely used for research and toxicity testing. In contrast, small ruminants, such as sheep and goats, are rarely used as animal models to evaluate toxicity in male reproductive organs. To the best of our knowledge, a comparable spermatogenic staging system in rams has not yet been fully characterised. Hence, this study aimed to adapt the existing spermatogenic staging based on acrosome development in bull testes to fit the seminiferous epithelium cycle of ram testes. The results show that spermatogenic staging based on acrosome development in bull testes can, with slight modifications, be efficiently used for the staging of ram testes.
Collapse
Affiliation(s)
- Sabina Šturm
- Institute of Pathology, Wild Animals, Fish and Bees, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, Ljubljana, 1000, Slovenia
| | - Tanja Švara
- Institute of Pathology, Wild Animals, Fish and Bees, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, Ljubljana, 1000, Slovenia
| | - Ellinor Spörndly-Nees
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Box 7011, 75007, Sweden
| | - Vesna Cerkvenik-Flajs
- Institute of Pathology, Wild Animals, Fish and Bees, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, Ljubljana, 1000, Slovenia
| | - Mitja Gombač
- Institute of Pathology, Wild Animals, Fish and Bees, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, Ljubljana, 1000, Slovenia
| | - Anna-Luisa Weber
- AnaPath Services GmbH, Hammerstrasse 49, Liestal, 4410, Switzerland
| | - Klaus Weber
- AnaPath Services GmbH, Hammerstrasse 49, Liestal, 4410, Switzerland
| |
Collapse
|
22
|
Raisis AL, Hosgood GL, Crawford N, Kästner S, Musk GC, Herrmann P, Mosing M. Comparison of pulmonary function in isoflurane anaesthetized ventilated sheep ( Ovis aries) following administration of intravenous xylazine versus medetomidine. Lab Anim 2021; 55:443-452. [PMID: 33645310 DOI: 10.1177/0023677220983366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Alpha2 receptor agonists (alpha2-agonists) are useful sedative and analgesic agents in sheep, but have adverse pulmonary effects, which are reportedly similar between different alpha2-agonists. This randomized crossover study compared pulmonary function after intravenous administration of an alpha2-agonist, either xylazine or an equipotent dose of medetomidine in 34 female sheep anaesthetized twice. Pulmonary function was assessed using spirometry, volumetric capnography, arterial blood gas analysis 1 min prior to, and 5 and 10 min after administration of the allocated alpha 2 agonist drug. Pulmonary structural changes were subsequently assessed using computed tomography (CT). Tachypnoea or hypoxaemia prompted reversal with atipamezole and exclusion of data. Data were analysed for a fixed effect of drug using a mixed effect linear model with significance set at p < 0.05. Ten sheep administered xylazine required atipamezole while none of sheep receiving medetomidine did. Xylazine produced significantly higher respiratory frequency, airway pressures, airway resistance and arterial carbon dioxide (CO2), and lower dynamic compliance, tidal volume, CO2 elimination and end tidal CO2 tension and arterial oxygen tension than medetomidine. This was associated with a significantly lower % of aerated tissue and higher % poorly and non-aerated tissue in CT images of sheep receiving xylazine versus medetomidine. In conclusion, xylazine administration produced marked decreases in pulmonary function, in ventilated isoflurane anaesthetized sheep, when compared to an equipotent dose of medetomidine when administered as an intravenous bolus supporting the use of medetomidine when alpha2-agonists are required.
Collapse
Affiliation(s)
- Anthea L Raisis
- School of Veterinary Medicine, Murdoch University, Australia
| | | | | | - Sabine Kästner
- Clinic for small domestic animals, University of Veterinary Medicine Hannover, Germany
| | - Gabrielle C Musk
- Animal Care Services, The University of Western Australia, Australia
| | - Peter Herrmann
- Department of Anaesthesiology, Intensive Care and Emergency Medicine, University Medical Centre Göttingen, Germany
| | - Martina Mosing
- School of Veterinary Medicine, Murdoch University, Australia
| |
Collapse
|
23
|
Kalds P, Luo Q, Sun K, Zhou S, Chen Y, Wang X. Trends towards revealing the genetic architecture of sheep tail patterning: Promising genes and investigatory pathways. Anim Genet 2021; 52:799-812. [PMID: 34472112 DOI: 10.1111/age.13133] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2021] [Indexed: 12/22/2022]
Abstract
Different sheep breeds have evolved after initial domestication, generating various tail phenotypic patterns. The phenotypic diversity of sheep tail patterns offers ideal materials for comparative analysis of its genetic basis. Evolutionary biologists, animal geneticists, breeders, and producers have been curious to clearly understand the underlying genetics behind phenotypic differences in sheep tails. Understanding the causal gene(s) and mutation(s) underlying these differences will help probe an evolutionary riddle, improve animal production performance, promote animal welfare, and provide lessons that help comprehend human diseases related to fat deposition (i.e., obesity). Historically, fat tails have served as an adaptive response to aridification and climate change. However, the fat tail is currently associated with compromised mating and animal locomotion, fat distribution in the animal body, increased raising costs, reduced consumer preference, and other animal welfare issues such as tail docking. The developing genomic approaches provide unprecedented opportunities to determine causal variants underlying phenotypic differences among populations. In the last decade, researchers have performed several genomic investigations to assess the genomic causality underlying phenotypic variations in sheep tails. Various genes have been suggested with the prominence of several potentially significant causatives, including the BMP2 and PDGFD genes associated with the fat tail phenotype and the TBXT gene linked with the caudal vertebrae number and tail length. Although the potential genes related to sheep tail characteristics have been revealed, the causal variant(s) and mutation(s) of these high-ranking candidate genes are still elusive and need further investigation. The review discusses the potential genes, sheds light on a knowledge gap, and provides possible investigative approaches that could help determine the specific genomic causatives of sheep tail patterns. Besides, characterizing and revealing the genetic determinism of sheep tails will help solve issues compromising sheep breeding and welfare in the future.
Collapse
Affiliation(s)
- P Kalds
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China.,Department of Animal and Poultry Production, Faculty of Environmental Agricultural Sciences, Arish University, El-Arish, Egypt
| | - Q Luo
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - K Sun
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - S Zhou
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Y Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - X Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
24
|
Brügger M, Démoulins T, Barut GT, Zumkehr B, Oliveira Esteves BI, Mehinagic K, Haas Q, Schögler A, Rameix-Welti MA, Eléouët JF, Moehrlen U, Marti TM, Schmid RA, Summerfield A, Posthaus H, Ruggli N, Hall SRR, Alves MP. Pulmonary mesenchymal stem cells are engaged in distinct steps of host response to respiratory syncytial virus infection. PLoS Pathog 2021; 17:e1009789. [PMID: 34320038 PMCID: PMC8351988 DOI: 10.1371/journal.ppat.1009789] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 08/09/2021] [Accepted: 07/08/2021] [Indexed: 02/06/2023] Open
Abstract
Lung-resident (LR) mesenchymal stem and stromal cells (MSCs) are key elements of the alveolar niche and fundamental regulators of homeostasis and regeneration. We interrogated their function during virus-induced lung injury using the highly prevalent respiratory syncytial virus (RSV) which causes severe outcomes in infants. We applied complementary approaches with primary pediatric LR-MSCs and a state-of-the-art model of human RSV infection in lamb. Remarkably, RSV-infection of pediatric LR-MSCs led to a robust activation, characterized by a strong antiviral and pro-inflammatory phenotype combined with mediators related to T cell function. In line with this, following in vivo infection, RSV invades and activates LR-MSCs, resulting in the expansion of the pulmonary MSC pool. Moreover, the global transcriptional response of LR-MSCs appears to follow RSV disease, switching from an early antiviral signature to repair mechanisms including differentiation, tissue remodeling, and angiogenesis. These findings demonstrate the involvement of LR-MSCs during virus-mediated acute lung injury and may have therapeutic implications.
Collapse
Affiliation(s)
- Melanie Brügger
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Thomas Démoulins
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - G. Tuba Barut
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Beatrice Zumkehr
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Blandina I. Oliveira Esteves
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Kemal Mehinagic
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Quentin Haas
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Aline Schögler
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Marie-Anne Rameix-Welti
- Université Paris-Saclay, INSERM, Université de Versailles St. Quentin, UMR 1173 (2I), Versailles, France
| | | | - Ueli Moehrlen
- Pediatric Surgery, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Thomas M. Marti
- Department of Biomedical Research, University of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ralph A. Schmid
- Department of Biomedical Research, University of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Artur Summerfield
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Horst Posthaus
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Nicolas Ruggli
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Sean R. R. Hall
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Gillies McIndoe Research Institute, Wellington, New Zealand
| | - Marco P. Alves
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
25
|
Capucciati A, Zucca FA, Monzani E, Zecca L, Casella L, Hofer T. Interaction of Neuromelanin with Xenobiotics and Consequences for Neurodegeneration; Promising Experimental Models. Antioxidants (Basel) 2021; 10:antiox10060824. [PMID: 34064062 PMCID: PMC8224073 DOI: 10.3390/antiox10060824] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 01/14/2023] Open
Abstract
Neuromelanin (NM) accumulates in catecholamine long-lived brain neurons that are lost in neurodegenerative diseases. NM is a complex substance made of melanic, peptide and lipid components. NM formation is a natural protective process since toxic endogenous metabolites are removed during its formation and as it binds excess metals and xenobiotics. However, disturbances of NM synthesis and function could be toxic. Here, we review recent knowledge on NM formation, toxic mechanisms involving NM, go over NM binding substances and suggest experimental models that can help identifying xenobiotic modulators of NM formation or function. Given the high likelihood of a central NM role in age-related human neurodegenerative diseases such as Parkinson’s and Alzheimer’s, resembling such diseases using animal models that do not form NM to a high degree, e.g., mice or rats, may not be optimal. Rather, use of animal models (i.e., sheep and goats) that better resemble human brain aging in terms of NM formation, as well as using human NM forming stem cellbased in vitro (e.g., mid-brain organoids) models can be more suitable. Toxicants could also be identified during chemical synthesis of NM in the test tube.
Collapse
Affiliation(s)
- Andrea Capucciati
- Department of Chemistry, University of Pavia, 27100 Pavia, Italy; (A.C.); (E.M.); (L.C.)
| | - Fabio A. Zucca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, 20054 Milan, Italy; (F.A.Z.); (L.Z.)
| | - Enrico Monzani
- Department of Chemistry, University of Pavia, 27100 Pavia, Italy; (A.C.); (E.M.); (L.C.)
| | - Luigi Zecca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, 20054 Milan, Italy; (F.A.Z.); (L.Z.)
| | - Luigi Casella
- Department of Chemistry, University of Pavia, 27100 Pavia, Italy; (A.C.); (E.M.); (L.C.)
| | - Tim Hofer
- Department of Environmental Health, Norwegian Institute of Public Health, P.O. Box 222 Skøyen, N-0213 Oslo, Norway
- Correspondence: ; Tel.: +47-21076671
| |
Collapse
|
26
|
The specific features of the developing T cell compartment of the neonatal lung are a determinant of respiratory syncytial virus immunopathogenesis. PLoS Pathog 2021; 17:e1009529. [PMID: 33909707 PMCID: PMC8109812 DOI: 10.1371/journal.ppat.1009529] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 05/10/2021] [Accepted: 04/05/2021] [Indexed: 11/19/2022] Open
Abstract
The human respiratory syncytial virus (RSV) is a major cause of severe lower respiratory tract infections in infants, possibly due to the properties of the immature neonatal pulmonary immune system. Using the newborn lamb, a classical model of human lung development and a translational model of RSV infection, we aimed to explore the role of cell-mediated immunity in RSV disease during early life. Remarkably, in healthy conditions, the developing T cell compartment of the neonatal lung showed major differences to that seen in the mature adult lung. The most striking observation being a high baseline frequency of bronchoalveolar IL-4-producing CD4+ and CD8+ T cells, which declined progressively over developmental age. RSV infection exacerbated this pro-type 2 environment in the bronchoalveolar space, rather than inducing a type 2 response per se. Moreover, regulatory T cell suppressive functions occurred very early to dampen this pro-type 2 environment, rather than shutting them down afterwards, while γδ T cells dropped and failed to produce IL-17. Importantly, RSV disease severity was related to the magnitude of those unconventional bronchoalveolar T cell responses. These findings provide novel insights in the mechanisms of RSV immunopathogenesis in early life, and constitute a major step for the understanding of RSV disease severity.
Collapse
|
27
|
Alvites RD, Branquinho MV, Sousa AC, Lopes B, Sousa P, Mendonça C, Atayde LM, Maurício AC. Small Ruminants and Its Use in Regenerative Medicine: Recent Works and Future Perspectives. BIOLOGY 2021; 10:biology10030249. [PMID: 33810087 PMCID: PMC8004958 DOI: 10.3390/biology10030249] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 12/16/2022]
Abstract
Simple Summary Small ruminants such as sheep and goats have been increasingly used as animal models due to their dimensions, physiology and anatomy identical to those of humans. Their low costs, ease of accommodation, great longevity and easy handling make them advantageous animals to be used in a wide range of research work. Although there is already a lot of scientific literature describing these species, their use still lacks some standardization. The purpose of this review is to summarize the general principles related to the use of small ruminants as animal models for scientific research. Abstract Medical and translational scientific research requires the use of animal models as an initial approach to the study of new therapies and treatments, but when the objective is an exploration of translational potentialities, classical models fail to adequately mimic problems in humans. Among the larger animal models that have been explored more intensely in recent decades, small ruminants, namely sheep and goats, have emerged as excellent options. The main advantages associated to the use of these animals in research works are related to their anatomy and dimensions, larger than conventional laboratory animals, but very similar to those of humans in most physiological systems, in addition to their low maintenance and feeding costs, tendency to be docile, long life expectancies and few ethical complications raised in society. The most obvious disadvantages are the significant differences in some systems such as the gastrointestinal, and the reduced amount of data that limits the comparison between works and the validation of the characterization essays. Despite everything, recently these species have been increasingly used as animal models for diseases in different systems, and the results obtained open doors for their more frequent and advantageous use in the future. The purpose of this review is to summarize the general principles related to the use of small ruminants as animal models, with a focus on regenerative medicine, to group the most relevant works and results published recently and to highlight the potentials for the near future in medical research.
Collapse
Affiliation(s)
- Rui Damásio Alvites
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Mariana Vieira Branquinho
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Ana Catarina Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Bruna Lopes
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Patrícia Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Carla Mendonça
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Luís Miguel Atayde
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Ana Colette Maurício
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
- Correspondence: ; Tel.: +351-919-071-286 or +351-220-428-000
| |
Collapse
|
28
|
Could cold plasma act synergistically with allogeneic mesenchymal stem cells to improve wound skin regeneration in a large size animal model? Res Vet Sci 2021; 136:97-110. [PMID: 33596495 DOI: 10.1016/j.rvsc.2021.01.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 01/05/2021] [Accepted: 01/24/2021] [Indexed: 02/06/2023]
Abstract
Skin wound healing may sometimes lead to open sores that persist for long periods and expensive hospitalization is needed. Among different kinds of therapeutic innovative approaches, mesenchymal stem cells (MSCs) and low-temperature atmospheric pressure cold plasma (ionized gas) have been recently tested to improve this regenerative process. To optimize wound healing the present study intended to combine, for the first time, these two novel approaches in a large size animal wound healing model with the aim of assessing the putative dual beneficial effects. Based on clinical, histopathological, and molecular results a synergistic action in a second intention healing wound in sheep has been observed. Experimental wounds treated with cold plasma and MSCs showed a slower but more effective healing compared to the single treatment, as observed in previous studies. The combined treatment improved the correct development of skin appendages and structural proteins of the dermis showing the potential of the dual combination as a safe and effective tool for skin regeneration in the veterinary clinical field.
Collapse
|
29
|
Sécher T, Bodier-Montagutelli E, Guillon A, Heuzé-Vourc'h N. Correlation and clinical relevance of animal models for inhaled pharmaceuticals and biopharmaceuticals. Adv Drug Deliv Rev 2020; 167:148-169. [PMID: 32645479 DOI: 10.1016/j.addr.2020.06.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/10/2020] [Accepted: 06/29/2020] [Indexed: 12/01/2022]
Abstract
Nonclinical studies are fundamental for the development of inhaled drugs, as for any drug product, and for successful translation to clinical practice. They include in silico, in vitro, ex vivo and in vivo studies and are intended to provide a comprehensive understanding of the inhaled drug beneficial and detrimental effects. To date, animal models cannot be circumvented during drug development programs, acting as surrogates of humans to predict inhaled drug response, fate and toxicity. Herein, we review the animal models used during the different development stages of inhaled pharmaceuticals and biopharmaceuticals, highlighting their strengths and limitations.
Collapse
Affiliation(s)
- T Sécher
- INSERM, Research Center for Respiratory Diseases, U1100, Tours, France; University of Tours, Tours, France
| | - E Bodier-Montagutelli
- INSERM, Research Center for Respiratory Diseases, U1100, Tours, France; University of Tours, Tours, France; CHRU de Tours, Pharmacy Department, Tours, France
| | - A Guillon
- INSERM, Research Center for Respiratory Diseases, U1100, Tours, France; University of Tours, Tours, France; CHRU de Tours, Critical Care Department, Tours, France
| | - N Heuzé-Vourc'h
- INSERM, Research Center for Respiratory Diseases, U1100, Tours, France; University of Tours, Tours, France.
| |
Collapse
|
30
|
Yiew XT, Bateman SW, Hahn RG, Bersenas AME, Muir WW. Understanding Volume Kinetics: The Role of Pharmacokinetic Modeling and Analysis in Fluid Therapy. Front Vet Sci 2020; 7:587106. [PMID: 33330713 PMCID: PMC7714761 DOI: 10.3389/fvets.2020.587106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/12/2020] [Indexed: 11/13/2022] Open
Abstract
Fluid therapy is a rapidly evolving yet imprecise clinical practice based upon broad assumptions, species-to-species extrapolations, obsolete experimental evidence, and individual preferences. Although widely recognized as a mainstay therapy in human and veterinary medicine, fluid therapy is not always benign and can cause significant harm through fluid overload, which increases patient morbidity and mortality. As with other pharmaceutical substances, fluids exert physiological effects when introduced into the body and therefore should be considered as "drugs." In human medicine, an innovative adaptation of pharmacokinetic analysis for intravenous fluids known as volume kinetics using serial hemoglobin dilution and urine output has been developed, refined, and investigated extensively for over two decades. Intravenous fluids can now be studied like pharmaceutical drugs, leading to improved understanding of their distribution, elimination, volume effect, efficacy, and half-life (duration of effect) under various physiologic conditions, making evidence-based approaches to fluid therapy possible. This review article introduces the basic concepts of volume kinetics, its current use in human and animal research, as well as its potential and limitations as a research tool for fluid therapy research in veterinary medicine. With limited evidence to support our current fluid administration practices in veterinary medicine, a greater understanding of volume kinetics and body water physiology in veterinary species would ideally provide some evidence-based support for safer and more effective intravenous fluid prescriptions in veterinary patients.
Collapse
Affiliation(s)
- Xiu Ting Yiew
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Shane W. Bateman
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Robert G. Hahn
- Research Unit, Södertälje Hospital, Södertälje, Sweden
- Karolinska Institutet, Danderyds Hospital (KIDS), Stockholm, Sweden
| | - Alexa M. E. Bersenas
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - William W. Muir
- College of Veterinary Medicine, Lincoln Memorial University, Harrogate, TN, United States
| |
Collapse
|
31
|
McBride GM, Soo JY, Varcoe T, Morrison JL, Wiese MD. Development of a method to determine cytochrome P450 1A2, 2C9, 2D6 and 3A4 activity sheep hepatic microsomes. J Pharmacol Toxicol Methods 2020; 106:106934. [PMID: 33080390 DOI: 10.1016/j.vascn.2020.106934] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 09/03/2020] [Accepted: 09/22/2020] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Ex vivo studies of human fetal hepatic drug metabolism are uncommon as it requires access to functional liver tissue and therefore raises practical and ethical concerns. Large animal models provide an alternative opportunity to study changes in cytochrome P450 (CYP) activity in the mother and fetus during pregnancy. We aimed to develop methods to determine the activity of CYP1A2, CYP2C9, CYP2D6 and CYP3A4 in sheep hepatic microsomes. METHODS We identified optimal conditions to determine the activity of CYP1A2 (using the probe drug phenacetin), CYP2C9 (diclofenac), CYP2D6 (dextromethorphan) and CYP3A4 (midazolam) by varying techniques for microsome extraction, probe drug concentration, incubation time and microsome concentration. The specificity of each probe drug was assessed by determining the rate of metabolism when specific CYP enzyme inhibitors were included in the reaction. RESULTS The optimum incubation time and probe drug concentration was six hours with 5 μM phenacetin (CYP1A2), four hours with 10 μM diclofenac (CYP2C9), 30 min with 1 μM of midazolam (CYP3A4) and 10 min with 1 μM dextromethorphan (CYP2D6). For both CYP2D6 and CYP3A4 reactions required 20 μg of microsomal protein, whereas for CYP1A2 and CYP2C9, reactions required 40 μg of microsomal protein. Metabolism of phenacetin, dextromethorphan and midazolam was reduced by specific enzyme inhibitors, but the specific CYP2C9 inhibitor sulfaphenazole did not substantially inhibit diclofenac metabolism. DISCUSSION This study identifies the optimal conditions for determining CYP activity in maternal sheep hepatic microsomes. In doing so, we have developed a standardised protocol for assessment of microsomal activity of CYP3A4, CYP1A2 and CYP2D6, but we were unable to optimise conditions for assessment of CYP2C9. This approach can be applied to investigate the impact of pregnancy complications on maternal and fetal hepatic drug metabolism.
Collapse
Affiliation(s)
- Grace M McBride
- Early Origins of Adult Health Research Group, Australia; UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Jia Yin Soo
- Early Origins of Adult Health Research Group, Australia; UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Tamara Varcoe
- Early Origins of Adult Health Research Group, Australia; UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Australia; UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Michael D Wiese
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia.
| |
Collapse
|
32
|
Wojtulewicz K, Krawczyńska A, Tomaszewska-Zaremba D, Wójcik M, Herman AP. Effect of Acute and Prolonged Inflammation on the Gene Expression of Proinflammatory Cytokines and Their Receptors in the Anterior Pituitary Gland of Ewes. Int J Mol Sci 2020; 21:E6939. [PMID: 32967383 PMCID: PMC7554822 DOI: 10.3390/ijms21186939] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/19/2020] [Accepted: 09/20/2020] [Indexed: 01/01/2023] Open
Abstract
An acute and prolonged inflammation inhibits the reproduction process by the disruption of the neurohormonal activity of the hypothalamic-pituitary-gonadal axis. It is thought that these changes may be caused by proinflammatory cytokines, i.e., interleukin (IL) -1β, IL-6 and tumor necrosis factor (TNF) α. The aim of this study was to determine the effect of an acute and prolonged inflammation on the expression of genes encoding cytokine and their receptors, gonadotropin releasing hormone receptor (GnRHR), beta subunits of luteinizing hormone (LHβ) and follicle-stimulating (FSHβ) in the anterior pituitary (AP). Moreover, the circulating concentration of LH and FSH was also assayed. Two experiments were carried out on adult ewes which were divided into two control groups and treated with lipopolysaccharide (LPS; 400 ng / kg). Acute inflammation was caused by a single injection of LPS into the external jugular vein, while the chronic inflammation was induced by seven times LPS injection (one a day). In both experiments, animals were euthanized 3h after the last LPS / NaCl injection and the blood samples collected 15 min before euthanasia. An acute inflammation stimulates the expression of the IL-1β, IL-6 and TNFα genes and their receptors in the AP of sheep. Prolonged inflammation increased TNFα gene expression and both types of TNFα and IL-6 receptors. Both an acute and prolonged inflammation inhibited LHβ gene expression in the AP and reduced LH level in blood. A sevenfold LPS injection raises FSH concentration. The gene expression of GnRHR was reduced in the ovine AP only after a single injection of endotoxin. Our results suggest that there are important differences in the way how an acute and prolonged inflammation influence proinflammatory cytokines and their receptors gene expression in the AP of anestrous ewes, which could be reflected by differences in the AP secretory activity during these states.
Collapse
Affiliation(s)
- Karolina Wojtulewicz
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 03-105 Jabłonna, Poland; (A.K.); (D.T.-Z.); (M.W.); (A.P.H.)
| | | | | | | | | |
Collapse
|
33
|
Rocha BR, Martins G, Lilenbaum W. An historical view of the experimental leptospiral infection in ruminants. Comp Immunol Microbiol Infect Dis 2020; 73:101532. [PMID: 32980802 DOI: 10.1016/j.cimid.2020.101532] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 08/13/2020] [Indexed: 10/23/2022]
Abstract
The first experimental infections with Leptospira in ruminants were conducted in the 1950s, primarily assessed the pathogenesis caused by serovar Pomona in cows. Throughout the decades, experimental infections have also demonstrated the clinical aspects of the infection by other strains, mainly Hardjo. Despite the important outcomes observed in experimental infections in ruminants, there is still a large discrepancy regarding the ideal dose, route, strain, model species or animal age that should be used to reproduce the acute and chronic leptospirosis in ruminants. In this context, the present study aimed to review the historical processes involved on the experimental leptospiral infection in ruminants. The inclusion criteria were papers that clearly described inoculation route, strain, dose, clinical signs and animal age. Overall, 37 experiments were noted. The most frequently reported clinical signs were fever, prostration, hematuria and death, with the majority of them occurring in young animals infected by incidental strains. Regarding reproductive problems, they occurred in the majority of the experiments and were also more related to incidental strains. In this context, abortions, retained placenta and weak fetuses were the most frequent symptoms. Noteworthy that although the mechanisms of the clinical acute disease either systemic or reproductive, is reasonably well understood, the physiopathology involved on reproductive problems due to the silent chronic infection is less discussed and remains to be elucidated. In this context, it is evident the need for studies focused on the genital infection and reproductive aspects of leptospiral infection in ruminants.
Collapse
Affiliation(s)
- Bruno Ribeiro Rocha
- Laboratório de Bacteriologia Veterinária, Departamento de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Gabriel Martins
- Laboratório de Bacteriologia Veterinária, Departamento de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil.
| | - Walter Lilenbaum
- Laboratório de Bacteriologia Veterinária, Departamento de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| |
Collapse
|
34
|
Smith JS, Mochel JP, Seo YJ, Ahrens AP, Griffith RW. Evaluation of a Pasteurella multocida Respiratory Disease Induction Model for Goats ( Capra aegagrus hircus). Comp Med 2020; 70:323-328. [PMID: 32907695 DOI: 10.30802/aalas-cm-20-000002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Infectious respiratory diseases are a serious health concern worldwide. However, few models describe the experimental induction of lung infection, or the effect of experimental infection on clinical pathologic parameters in goats. Goats offer benefits compared to cattle because of size and tractability and compared to sheep with regard to specific features of their anatomy. In previous experimental models of infection in goats, coadministration of an immunosuppressive dose of a corticosteroid is common; however, protocols that use corticosteroid often note mortality as an adverse effect. We therefore investigated an infection protocol that did not use immunosuppression but instead relied on 2 intratracheal inoculations of Pasteurella multocida in healthy meat goats to induce clinical and hematologic changes associated with respiratory infection. Healthy Boer or Boer-Kiko cross goats (n = 6; age, 10 mo) were inoculated with Pasteurella multocida and were monitored over a 312-h period for clinical and hematologic parameters of infection. After induction of pneumonia, the goats had a significant 1.2 °C rise in rectal temperature and auscultatable rales for up to 96 h. Lymphocyte counts, serum amyloid A values, and respiratory scores were significantly different before and after induction of disease and were consistent with respiratory infection. No mortality was associated with this experimental infection, and minimal gross pathologic changes were noted at study termination. The clinical and pathologic findings of this study suggest a potentially reproducible method of establishing clinical respiratory infection in goats. The repeated intratracheal inoculation method of inducing caprine respiratory disease can be used to produce experimental respiratory disease in goats when the use of corticosteroid is not desirable. With the feasibility of this method established, additional research evaluating the optimal dose and frequency of P. multocida administration is needed.
Collapse
Affiliation(s)
- Joe S Smith
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, Iowa; Systems Modelling and Reverse Translational Pharmacology, Iowa State University, Ames, Iowa; Biomedical Sciences, Iowa State University, Ames, Iowa; Current affiliation. Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee;,
| | - Jonathan P Mochel
- Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, Iowa; Systems Modelling and Reverse Translational Pharmacology, Iowa State University, Ames, Iowa; Biomedical Sciences, Iowa State University, Ames, Iowa
| | - Yeon-Jung Seo
- Systems Modelling and Reverse Translational Pharmacology, Iowa State University, Ames, Iowa; Biomedical Sciences, Iowa State University, Ames, Iowa
| | - Amanda P Ahrens
- Laboratory Animal Resources, Iowa State University, Ames, Iowa
| | - Ronald W Griffith
- Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, Iowa
| |
Collapse
|
35
|
Development and validation of ELISAs for the quantitation of interleukin (IL)-1β, IL-6, IL-8 and IL-10 in ovine plasma. J Immunol Methods 2020; 486:112835. [PMID: 32828792 DOI: 10.1016/j.jim.2020.112835] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 08/02/2020] [Accepted: 08/12/2020] [Indexed: 02/08/2023]
Abstract
There is growing evidence that inflammation underpins many common diseases. Inflammatory/immunomodulatory/immune mediators, such as cytokines, are key modulators of inflammation and mediate both immune cell recruitment and complex intracellular signalling pathways. Ovine models of disease are increasingly utilized in pre-clinical research, however existing methods for measuring cytokine levels are limited. We established and validated enzyme-linked immunosorbent assays (ELISAs) targeting interleukin (IL)-1β, IL-6, IL-8 and IL-10 in sheep plasma. These ELISAs showed high sensitivity and specificity with intra- and inter-assay CV's below 10%, and recovery rates between 82 and 123%. Sensitivity for IL-1β, IL-6, IL-8 and IL-10 were 117.6 pg/mL, 443.1 pg/mL, 30.9 pg/mL, and 64.3 pg/mL, respectively. ELISA test result reproducibility decreased significantly after 12 weeks of plasma storage at -80 °C. Therefore, for accurate cytokine measurements, plasma samples need to be tested within three months of sample collection to account for cytokine protein degradation. These ELISAs offer a reliable and convenient method to identify inflammatory cytokine changes in sheep, allowing key insights into the disease pathogenesis of these ruminants.
Collapse
|
36
|
Ribitsch I, Baptista PM, Lange-Consiglio A, Melotti L, Patruno M, Jenner F, Schnabl-Feichter E, Dutton LC, Connolly DJ, van Steenbeek FG, Dudhia J, Penning LC. Large Animal Models in Regenerative Medicine and Tissue Engineering: To Do or Not to Do. Front Bioeng Biotechnol 2020; 8:972. [PMID: 32903631 PMCID: PMC7438731 DOI: 10.3389/fbioe.2020.00972] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 07/27/2020] [Indexed: 12/13/2022] Open
Abstract
Rapid developments in Regenerative Medicine and Tissue Engineering has witnessed an increasing drive toward clinical translation of breakthrough technologies. However, the progression of promising preclinical data to achieve successful clinical market authorisation remains a bottleneck. One hurdle for progress to the clinic is the transition from small animal research to advanced preclinical studies in large animals to test safety and efficacy of products. Notwithstanding this, to draw meaningful and reliable conclusions from animal experiments it is critical that the species and disease model of choice is relevant to answer the research question as well as the clinical problem. Selecting the most appropriate animal model requires in-depth knowledge of specific species and breeds to ascertain the adequacy of the model and outcome measures that closely mirror the clinical situation. Traditional reductionist approaches in animal experiments, which often do not sufficiently reflect the studied disease, are still the norm and can result in a disconnect in outcomes observed between animal studies and clinical trials. To address these concerns a reconsideration in approach will be required. This should include a stepwise approach using in vitro and ex vivo experiments as well as in silico modeling to minimize the need for in vivo studies for screening and early development studies, followed by large animal models which more closely resemble human disease. Naturally occurring, or spontaneous diseases in large animals remain a largely untapped resource, and given the similarities in pathophysiology to humans they not only allow for studying new treatment strategies but also disease etiology and prevention. Naturally occurring disease models, particularly for longer lived large animal species, allow for studying disorders at an age when the disease is most prevalent. As these diseases are usually also a concern in the chosen veterinary species they would be beneficiaries of newly developed therapies. Improved awareness of the progress in animal models is mutually beneficial for animals, researchers, human and veterinary patients. In this overview we describe advantages and disadvantages of various animal models including domesticated and companion animals used in regenerative medicine and tissue engineering to provide an informed choice of disease-relevant animal models.
Collapse
Affiliation(s)
- Iris Ribitsch
- Veterm, Department for Companion Animals and Horses, University Equine Hospital, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Pedro M. Baptista
- Laboratory of Organ Bioengineering and Regenerative Medicine, Health Research Institute of Aragon (IIS Aragon), Zaragoza, Spain
| | - Anna Lange-Consiglio
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy
| | - Luca Melotti
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua, Italy
| | - Marco Patruno
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua, Italy
| | - Florien Jenner
- Veterm, Department for Companion Animals and Horses, University Equine Hospital, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Eva Schnabl-Feichter
- Clinical Unit of Small Animal Surgery, Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Luke C. Dutton
- Department of Clinical Sciences and Services, Royal Veterinary College, Hertfordshire, United Kingdom
| | - David J. Connolly
- Clinical Unit of Small Animal Surgery, Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Frank G. van Steenbeek
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Jayesh Dudhia
- Department of Clinical Sciences and Services, Royal Veterinary College, Hertfordshire, United Kingdom
| | - Louis C. Penning
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
37
|
Sitthicharoenchai P, Alnajjar S, Ackermann MR. A model of respiratory syncytial virus (RSV) infection of infants in newborn lambs. Cell Tissue Res 2020; 380:313-324. [PMID: 32347384 PMCID: PMC7223741 DOI: 10.1007/s00441-020-03213-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 04/01/2020] [Indexed: 12/29/2022]
Abstract
Many animal models have been established for respiratory syncytial virus (RSV) infection of infants with the purpose of studying the pathogenesis, immunological response, and pharmaceutical testing and the objective of finding novel therapies and preventive measures. This review centers on a neonatal lamb model of RSV infection that has similarities to RSV infection of infants. It includes a comprehensive description of anatomical and immunological similarities between ovine and human lungs along with comparison of pulmonary changes and immune responses with RSV infection. These features make the newborn lamb an effective model for investigating key aspects of RSV infection in infants. The importance of RSV lamb model application in preclinical therapeutic trials and current updates on new studies with the RSV-infected neonatal lamb are also highlighted.
Collapse
Affiliation(s)
- Panchan Sitthicharoenchai
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA USA
| | - Sarhad Alnajjar
- Department of Veterinary Pathology, College of Veterinary Medicine, University of Baghdad, Baghdad, Iraq
- LambCure LLC, Corvallis, OR USA
| | - Mark R. Ackermann
- LambCure LLC, Corvallis, OR USA
- Department of Biomedical Sciences and Oregon Veterinary Diagnostic Laboratory, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR USA
| |
Collapse
|
38
|
Fukuda S, Niimi Y, Andersen CR, Manyeza ER, Rojas JD, Prough DS, Enkhbaatar P. Blood carboxyhemoglobin elimination curve, half-lifetime, and arterial-venous differences in acute phase of carbon monoxide poisoning in ovine smoke inhalation injury model. Biochem Biophys Res Commun 2020; 526:141-146. [PMID: 32199614 DOI: 10.1016/j.bbrc.2020.03.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 03/08/2020] [Indexed: 10/24/2022]
Abstract
Smoke inhalation injury (SII) affects more than 50,000 people annually causing carbon monoxide (CO) poisoning. Although the increased blood level of carboxyhemoglobin (CO-Hb) is frequently used to confirm the diagnosis of SII, knowledge of its elimination in the acute phase is still limited. The aim of this study is to determine CO-Hb elimination rates and their differences in arterial (aCO-Hb) and mixed-venous (vCO-Hb) blood following severe SII in a clinically relevant ovine model. Forty-three chronically instrumented female sheep were subjected to SII (12 breaths, 4 sets) through tracheostomy tube under anesthesia and analgesia. After the SII, sheep were awakened and placed on a mechanical ventilator (FiO2 = 1.0, tidal volume 12 mL/kg, and PEEP = 5cmH2O) and monitored. Arterial and mixed-venous blood samples were withdrawn simultaneously for blood gas analysis at various time points to determine CO-HB half-lifetime and an elimination curve. The mean of highest aCO-Hb level during SII was 70.8 ± 13.9%. The aCO-Hb elimination curve showed an approximated exponential decay during the first 60 min. Per mixed linear regression model analysis, aCO-Hb significantly (p < 0.001) declined (4.3%/minute) with a decay constant lambda of 0.044. With this lambda, mean lifetime and half-lifetime of aCO-Hb were 22.7 and 15.7 min, respectively. The aCO-Hb was significantly lower compared to vCO-Hb at all-time points (0-180 min). To our knowledge, this is the first report describing CO-Hb elimination curve in the acute phase after severe SII in the clinically relevant ovine model. Our data shows that CO-Hb is decreasing in linear manner with supportive mechanical ventilation (0-60 min). The results may help to understand CO-Hb elimination curve in the acute phase and improvement of pre-hospital and initial clinical care in patients with CO poisoning.
Collapse
Affiliation(s)
- Satoshi Fukuda
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Yosuke Niimi
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA; Department of Plastic and Reconstructive Surgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Clark R Andersen
- Department of Preventive Medicine & Community Health, Office of Biostatistics, University of Texas Medical Branch, Galveston, TX, USA
| | - Ennert R Manyeza
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jose D Rojas
- Department of Respiratory Care, School of Health Professions, University of Texas Medical Branch, Galveston, TX, USA
| | - Donald S Prough
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Perenlei Enkhbaatar
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
39
|
Schwarz ER, Oliveira LJ, Bonfante F, Pu R, Pozor MA, Maclachlan NJ, Beachboard S, Barr KL, Long MT. Experimental Infection of Mid-Gestation Pregnant Female and Intact Male Sheep with Zika Virus. Viruses 2020; 12:v12030291. [PMID: 32156037 PMCID: PMC7150993 DOI: 10.3390/v12030291] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/24/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022] Open
Abstract
Zika virus (ZIKV) is an arbovirus that causes birth defects, persistent male infection, and sexual transmission in humans. The purpose of this study was to continue the development of an ovine ZIKV infection model; thus, two experiments were undertaken. In the first experiment, we built on previous pregnant sheep experiments by developing a mid-gestation model of ZIKV infection. Four pregnant sheep were challenged with ZIKV at 57–64 days gestation; two animals served as controls. After 13–15 days (corresponding with 70–79 days of gestation), one control and two infected animals were euthanized; the remaining animals were euthanized at 20–22 days post-infection (corresponding with 77–86 days of gestation). In the second experiment, six sexually mature, intact, male sheep were challenged with ZIKV and two animals served as controls. Infected animals were serially euthanized on days 2–6 and day 9 post-infection with the goal of isolating ZIKV from the male reproductive tract. In the mid-gestation study, virus was detected in maternal placenta and spleen, and in fetal organs, including the brains, spleens/liver, and umbilicus of infected fetuses. Fetuses from infected animals had visibly misshapen heads and morphometrics revealed significantly smaller head sizes in infected fetuses when compared to controls. Placental pathology was evident in infected dams. In the male experiment, ZIKV was detected in the spleen, liver, testes/epididymides, and accessory sex glands of infected animals. Results from both experiments indicate that mid-gestation ewes can be infected with ZIKV with subsequent disruption of fetal development and that intact male sheep are susceptible to ZIKV infection and viral dissemination and replication occurs in highly vascular tissues (including those of the male reproductive tract).
Collapse
Affiliation(s)
- Erika R. Schwarz
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA; (E.R.S.); (R.P.); (S.B.)
| | - Lilian J. Oliveira
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA;
| | - Francesco Bonfante
- Laboratory of Experimental Animal Models, Division of Comparative Biomedical Sciences, Instituto Zooprofilattico Sperimentale delle Venezie, 35020 Legnaro, Italy;
| | - Ruiyu Pu
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA; (E.R.S.); (R.P.); (S.B.)
| | - Malgorzata A. Pozor
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA;
| | - N. James Maclachlan
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA;
| | - Sarah Beachboard
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA; (E.R.S.); (R.P.); (S.B.)
| | - Kelli L. Barr
- Department of Biology, College of Arts and Sciences, Baylor University, Waco, TX 76798, USA;
| | - Maureen T. Long
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA; (E.R.S.); (R.P.); (S.B.)
- Correspondence:
| |
Collapse
|
40
|
Comparative Therapeutic Potential of ALX-0171 and Palivizumab against Respiratory Syncytial Virus Clinical Isolate Infection of Well-Differentiated Primary Pediatric Bronchial Epithelial Cell Cultures. Antimicrob Agents Chemother 2020; 64:AAC.02034-19. [PMID: 31767728 DOI: 10.1128/aac.02034-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 11/20/2019] [Indexed: 01/09/2023] Open
Abstract
Respiratory syncytial virus (RSV) causes severe lower respiratory tract infections in young infants. There are no RSV-specific treatments available. Ablynx has been developing an anti-RSV F-specific nanobody, ALX-0171. To characterize the therapeutic potential of ALX-0171, we exploited our well-differentiated primary pediatric bronchial epithelial cell (WD-PBEC)/RSV infection model, which replicates several hallmarks of RSV disease in vivo Using 2 clinical isolates (BT2a and Memphis 37), we compared the therapeutic potential of ALX-0171 with that of palivizumab, which is currently prescribed for RSV prophylaxis in high-risk infants. ALX-0171 treatment (900 nM) at 24 h postinfection reduced apically released RSV titers to near or below the limit of detection within 24 h for both strains. Progressively lower doses resulted in concomitantly diminished RSV neutralization. ALX-0171 was approximately 3-fold more potent in this therapeutic RSV/WD-PBEC model than palivizumab (mean 50% inhibitory concentration [IC50] = 346.9 to 363.6 nM and 1,048 to 1,090 nM for ALX-0171 and palivizumab, respectively), irrespective of the clinical isolate. The number of viral genomic copies (GC) was determined by quantitative reverse transcription-PCR (RT-qPCR), and the therapeutic effect of ALX-0171 treatment at 300 and 900 nM was found to be considerably lower and the number of GCs reduced only moderately (0.62 to 1.28 log10 copies/ml). Similar findings were evident for palivizumab. Therefore, ALX-0171 was very potent at neutralizing RSV released from apical surfaces but had only a limited impact on virus replication. The data indicate a clear disparity between viable virus neutralization and GC viral load, the latter of which does not discriminate between viable and neutralized RSV. This report validates the RSV/WD-PBEC model for the preclinical evaluation of RSV antivirals.
Collapse
|
41
|
Abstract
Asthma is a heterogeneous inflammatory disease of the airways that is associated with airway hyperresponsiveness and airflow limitation. Although asthma was once simply categorized as atopic or nonatopic, emerging analyses over the last few decades have revealed a variety of asthma endotypes that are attributed to numerous pathophysiological mechanisms. The classification of asthma by endotype is primarily routed in different profiles of airway inflammation that contribute to bronchoconstriction. Many asthma therapeutics target G protein-coupled receptors (GPCRs), which either enhance bronchodilation or prevent bronchoconstriction. Short-acting and long-acting β 2-agonists are widely used bronchodilators that signal through the activation of the β 2-adrenergic receptor. Short-acting and long-acting antagonists of muscarinic acetylcholine receptors are used to reduce bronchoconstriction by blocking the action of acetylcholine. Leukotriene antagonists that block the signaling of cysteinyl leukotriene receptor 1 are used as an add-on therapy to reduce bronchoconstriction and inflammation induced by cysteinyl leukotrienes. A number of GPCR-targeting asthma drug candidates are also in different stages of development. Among them, antagonists of prostaglandin D2 receptor 2 have advanced into phase III clinical trials. Others, including antagonists of the adenosine A2B receptor and the histamine H4 receptor, are in early stages of clinical investigation. In the past decade, significant research advancements in pharmacology, cell biology, structural biology, and molecular physiology have greatly deepened our understanding of the therapeutic roles of GPCRs in asthma and drug action on these GPCRs. This review summarizes our current understanding of GPCR signaling and pharmacology in the context of asthma treatment. SIGNIFICANCE STATEMENT: Although current treatment methods for asthma are effective for a majority of asthma patients, there are still a large number of patients with poorly controlled asthma who may experience asthma exacerbations. This review summarizes current asthma treatment methods and our understanding of signaling and pharmacology of G protein-coupled receptors (GPCRs) in asthma therapy, and discusses controversies regarding the use of GPCR drugs and new opportunities in developing GPCR-targeting therapeutics for the treatment of asthma.
Collapse
Affiliation(s)
- Stacy Gelhaus Wendell
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (S.G.W., C.Z.); Bioinformatics Institute, Agency for Science, Technology, and Research, Singapore (H.F.); and Department of Biological Sciences, National University of Singapore, and Center for Computational Biology, DUKE-NUS Medical School, Singapore (H.F.)
| | - Hao Fan
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (S.G.W., C.Z.); Bioinformatics Institute, Agency for Science, Technology, and Research, Singapore (H.F.); and Department of Biological Sciences, National University of Singapore, and Center for Computational Biology, DUKE-NUS Medical School, Singapore (H.F.)
| | - Cheng Zhang
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (S.G.W., C.Z.); Bioinformatics Institute, Agency for Science, Technology, and Research, Singapore (H.F.); and Department of Biological Sciences, National University of Singapore, and Center for Computational Biology, DUKE-NUS Medical School, Singapore (H.F.)
| |
Collapse
|
42
|
George R, Ma A, Motyka B, Shi YE, Liu Q, Griebel P. A dendritic cell-targeted chimeric hepatitis B virus immunotherapeutic vaccine induces both cellular and humoral immune responses in vivo. Hum Vaccin Immunother 2019; 16:779-792. [PMID: 31687875 PMCID: PMC7227651 DOI: 10.1080/21645515.2019.1689081] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Chimigen® HBV Immunotherapeutic Vaccine (C-HBV), a recombinant chimeric fusion protein comprising hepatitis B virus (HBV) S1 and S2 surface antigen fragments, Core antigen and a murine monoclonal antibody heavy chain fragment (Fc), was designed and produced in Sf9 insect cells. C-HBV targets the host immune system through specific receptors present on dendritic cells (DCs) which facilitates antigen internalization, processing, and presentation on MHC class I and II to induce both cellular and humoral immune responses against HBV antigens. T cell responses, previously assessed by ex vivo antigen presentation assays using human peripheral blood mononuclear cell (PBMC)-derived DCs and T cells from uninfected and HBV chronic-infected donors, demonstrated that C-HBV was highly immunogenic. A vaccine dose response study was performed in sheep to analyze the immunogenicity of C-HBV in vivo. Sheep (n = 8/group) received three consecutive subcutaneous injections of each dose of C-HBV at four-week intervals. Analysis of serum antibody levels confirmed C-HBV induced a dose-dependent antibody response to C-HBV and S1/S2-Core. Kinetics of the S1/S2-Core specific antibody response was similar to hepatitis B surface antigen (HBsAg)-specific antibody responses induced by ENGERIX-B. Analysis of cell-mediated immune responses (CMI) confirmed C-HBV induced both dose-dependent S1/S2-Core-specific lymphocyte proliferative responses and IFN-γ secretion. These responses were stronger with blood lymphocytes than with cells isolated from the lymph node draining the vaccination site. No correlation was seen between antibody titers and CMI. The results confirm C-HBV is an effective delivery vehicle for the induction of T cell responses and may be an appropriate candidate for immunotherapy for chronic HBV infections.
Collapse
Affiliation(s)
| | - Allan Ma
- Akshaya Bio Inc., Edmonton, Alberta, Canada
| | - Bruce Motyka
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Yuenian Eric Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiang Liu
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, SK, Canada.,School of Public Health, University of Saskatchewan, Saskatoon, SK, Canada.,Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Philip Griebel
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, SK, Canada.,School of Public Health, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
43
|
Dewage SNV, Organ L, Koumoundouros E, Derseh HB, Perera KUE, Samuel CS, Stent AW, Snibson KJ. The efficacy of pirfenidone in a sheep model of pulmonary fibrosis. Exp Lung Res 2019; 45:310-322. [PMID: 31762329 DOI: 10.1080/01902148.2019.1695019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive fibrotic lung disease with unknown cause. While the drugs nintedanib and pirfenidone have been approved for the treatment of IPF, they only slow disease progression and can induce several side-effects, suggesting that there is still an unmet need to develop new efficacious drugs, and interventions strategies, to combat this disease. We have recently developed a sheep model of pulmonary fibrosis for the preclinical testing of novel anti-fibrotic drugs. The aim of this study was to assess the effects of pirfenidone to ascertain its suitability as a benchmark for comparing other novel therapeutics in this sheep model. To initiate localized fibrosis, sheep were given two infusions of bleomycin (0.6 U/ml per infusion), a fortnight apart, to a specific lung segment. The contralateral lung segment in each sheep was infused with saline to act as an internal control. Two weeks after the final bleomycin infusion, either pirfenidone or methylcellulose (vehicle control) were administered orally to sheep twice daily for 5 weeks. Results showed that sheep treated with pirfenidone had improved lung function, ameliorated fibrotic pathology, lower numbers of active myofibroblasts, and reduced extra cellular matrix deposition when compared with the relevant measurements obtained from control sheep treated with vehicle. This study showed that pirfenidone can attenuate bleomycin-induced pulmonary fibrosis in sheep, and can therefore be used as a positive control to assess other novel therapeutics for IPF in this model.
Collapse
Affiliation(s)
- Sasika N V Dewage
- Faculty of Veterinary and Agricultural Science, The University of Melbourne, Parkville, Australia
| | - Louise Organ
- Nottingham Respiratory Research Unit, University of Nottingham, Nottingham, UK
| | - Emmanuel Koumoundouros
- Department of Electrical and Electronic Engineering, The University of Melbourne, Parkville, Australia
| | - Habtamu B Derseh
- Faculty of Veterinary and Agricultural Science, The University of Melbourne, Parkville, Australia
| | | | | | - Andrew W Stent
- Faculty of Veterinary Science and Agricultural Science, The University of Melbourne, Werribee, Australia
| | - Ken J Snibson
- Faculty of Veterinary and Agricultural Science, The University of Melbourne, Parkville, Australia
| |
Collapse
|
44
|
Stillman MW, Whittaker AL. Use and Efficacy of Analgesic Agents in Sheep ( Ovis aries) Used in Biomedical Research. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE 2019; 58:755-766. [PMID: 31604483 DOI: 10.30802/aalas-jaalas-19-000036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Sheep (Ovis aries) are widely used as large animal models in biomedical research. However, current literature on the use of analgesics in sheep generally focuses on an industry or farm level of use. This structured review evaluates use and efficacy of analgesics administered to sheep in a biomedical research setting. Electronic databases were searched with terms related to analgesia in research sheep. After application of exclusion criteria, 29 peer-reviewed publications were evaluated from 1995 to 2018. Drugs used for analgesia in sheep include opioids, α₂ agonists, NSAID, local anesthetics, NMDA receptor antagonists, and calcium channel blockers. Opioid agonists have previously been considered short acting and of questionable efficacy in sheep, but newer modalities may provide effective analgesia. NSAID may exhibit an analgesic effect only when inflammatory pain is present and may not be beneficial for use in acute pain models. α₂ agonists provide effective yet short-lived analgesia; however, side effects are of concern. Local anesthetics were previously widely used as stand-alone agents, as alternatives to the use of general anesthetics in sheep. These agents have since fallen out of favor as sole agents. Despite this, they provide a valuable analgesic effect when used as adjuncts to general anesthetic regimes. The NMDA antagonist ketamine provided good analgesia and is likely underutilized as an analgesic agent in sheep. Future controlled studies should further evaluate the analgesic properties of ketamine in sheep.
Collapse
Affiliation(s)
- Mark W Stillman
- School of Animal and Veterinary Sciences, University of Adelaide, Adelaide, Australia
| | - Alexandra L Whittaker
- School of Animal and Veterinary Sciences, University of Adelaide, Adelaide, Australia;,
| |
Collapse
|
45
|
da Rosa MC, Martins G, Rocha BR, Correia L, Ferronato G, Lilenbaum W, Dellagostin OA. Assessment of the immunogenicity of the leptospiral LipL32, LigAni, and LigBrep recombinant proteins in the sheep model. Comp Immunol Microbiol Infect Dis 2019; 65:176-180. [PMID: 31300110 DOI: 10.1016/j.cimid.2019.05.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 12/11/2022]
Abstract
Veterinary leptospirosis vaccines are composed of bacterins and present limitations, for example, the need for bacteriological culture and serovar-dependent immunity. Recombinant antigens represent a promising alternative. LigAni, LigBrep, and LipL32 proteins have been shown to promote a protective immune response against the homologous challenge in hamsters. Therefore, the next step is to evaluate the immunological properties of these immunogens in the actual hosts, as ruminants, which has never been performed before. The objective of this study was to evaluate the immunogenicity and potential adverse effects of the recombinant proteins LigAni, LigBrep, and LipL32 in the ovine model. For this, 16 Santa Inês sheep were allocated into three groups: two experimental (Groups A and B) and one control group (Group C). Group A was inoculated with a formulation containing the recombinant proteins in combination with the aluminum hydroxide adjuvant; Group B was inoculated with a formulation containing the recombinant proteins in combination with the Montanide adjuvant; and Group C was inoculated with adjuvants only. The results revealed that formulations containing the recombinant proteins induced total IgG seroconversion and led to a significant increase in antibody titers in the sheep model. Besides, there were no clinical changes or adverse effects. Thus, LigAni, LigBrep, and LipL32 proteins elicited a significant humoral immune response with elevated serum IgG levels, demonstrating that they possess the immunogenic and safety characteristics necessary to sustain their potential use as leptospirosis vaccines in the ruminant model.
Collapse
Affiliation(s)
- Matheus Costa da Rosa
- Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Gabriel Martins
- Laboratório de Bacteriologia Veterinária, Departamento de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Bruno Ribeiro Rocha
- Laboratório de Bacteriologia Veterinária, Departamento de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Lucas Correia
- Laboratório de Bacteriologia Veterinária, Departamento de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Giuliana Ferronato
- Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Walter Lilenbaum
- Laboratório de Bacteriologia Veterinária, Departamento de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Odir Antônio Dellagostin
- Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia Universidade Federal de Pelotas, Pelotas, Rio Grande do Sul, Brazil.
| |
Collapse
|
46
|
Altamirano-Lagos MJ, Díaz FE, Mansilla MA, Rivera-Pérez D, Soto D, McGill JL, Vasquez AE, Kalergis AM. Current Animal Models for Understanding the Pathology Caused by the Respiratory Syncytial Virus. Front Microbiol 2019; 10:873. [PMID: 31130923 PMCID: PMC6510261 DOI: 10.3389/fmicb.2019.00873] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 04/04/2019] [Indexed: 12/14/2022] Open
Abstract
The human respiratory syncytial virus (hRSV) is the main etiologic agent of severe lower respiratory tract infections that affect young children throughout the world, associated with significant morbidity and mortality, becoming a serious public health problem globally. Up to date, no licensed vaccines are available to prevent severe hRSV-induced disease, and the generation of safe-effective vaccines has been a challenging task, requiring constant biomedical research aimed to overcome this ailment. Among the difficulties presented by the study of this pathogen, it arises the fact that there is no single animal model that resembles all aspects of the human pathology, which is due to the specificity that this pathogen has for the human host. Thus, for the study of hRSV, different animal models might be employed, depending on the goal of the study. Of all the existing models, the murine model has been the most frequent model of choice for biomedical studies worldwide and has been of great importance at contributing to the development and understanding of vaccines and therapies against hRSV. The most notable use of the murine model is that it is very useful as a first approach in the development of vaccines or therapies such as monoclonal antibodies, suggesting in this way the direction that research could have in other preclinical models that have higher maintenance costs and more complex requirements in its management. However, several additional different models for studying hRSV, such as other rodents, mustelids, ruminants, and non-human primates, have been explored, offering advantages over the murine model. In this review, we discuss the various applications of animal models to the study of hRSV-induced disease and the advantages and disadvantages of each model, highlighting the potential of each model to elucidate different features of the pathology caused by the hRSV infection.
Collapse
Affiliation(s)
- María José Altamirano-Lagos
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fabián E. Díaz
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Miguel Andrés Mansilla
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela Rivera-Pérez
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniel Soto
- Sección Biotecnología, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Jodi L. McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| | - Abel E. Vasquez
- Sección Biotecnología, Instituto de Salud Pública de Chile, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago, Chile
| | - Alexis M. Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
47
|
Larios Mora A, Detalle L, Gallup JM, Van Geelen A, Stohr T, Duprez L, Ackermann MR. Delivery of ALX-0171 by inhalation greatly reduces respiratory syncytial virus disease in newborn lambs. MAbs 2019; 10:778-795. [PMID: 29733750 PMCID: PMC6150622 DOI: 10.1080/19420862.2018.1470727] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a common cause of acute lower respiratory disease in infants and young children worldwide. Currently, treatment is supportive and no vaccines are available. The use of newborn lambs to model hRSV infection in human infants may provide a valuable tool to assess safety and efficacy of new antiviral drugs and vaccines. ALX-0171 is a trivalent Nanobody targeting the hRSV fusion (F) protein and its therapeutic potential was evaluated in newborn lambs infected with a human strain of RSV followed by daily ALX-0171 nebulization for 3 or 5 consecutive days. Colostrum-deprived newborn lambs were infected with hRSV-M37 before being treated by daily nebulization with either ALX-0171 or placebo. Two different treatment regimens were examined: day 1–5 or day 3–5 post-infection. Lambs were monitored daily for general well-being and clinical parameters. Respiratory tissues and bronchoalveolar lavage fluid were collected at day 6 post-inoculation for the quantification of viral lesions, lung viral titers, viral antigen and lung histopathology. Administration by inhalation of ALX-0171 was well-tolerated in these hRSV-infected newborn lambs. Robust antiviral effects and positive effects on hRSV-induced lung lesions and reduction in symptoms of illness were noted. These effects were still apparent when treatment start was delayed and coincided with peak viral loads (day 3 post-infection) and at a time point when signs of RSV disease were apparent. The latter design is expected to have high translational value for planned clinical trials. These results are indicative of the therapeutic potential of ALX-0171 in infants.
Collapse
Affiliation(s)
- Alejandro Larios Mora
- a College of Veterinary Medicine, Department of Veterinary Pathology , Iowa State University , Ames , IA , USA
| | | | - Jack M Gallup
- a College of Veterinary Medicine, Department of Veterinary Pathology , Iowa State University , Ames , IA , USA
| | - Albert Van Geelen
- a College of Veterinary Medicine, Department of Veterinary Pathology , Iowa State University , Ames , IA , USA
| | | | | | - Mark R Ackermann
- a College of Veterinary Medicine, Department of Veterinary Pathology , Iowa State University , Ames , IA , USA
| |
Collapse
|
48
|
Dunbar ML, Walkowiak KJ, Faustich JS, Rendahl AK, Graham ML. Preliminary Evaluation of Sustained-release Compared with Conventional Formulations of Meloxicam in Sheep ( Ovis aries). JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE 2019; 58:339-345. [PMID: 30846026 DOI: 10.30802/aalas-jaalas-18-000076] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Sustained-release (SR) drugs refine current analgesic regimens by alleviating the need for multiple sessions of handling and restraint and by reducing the local tissue irritation that can occur due to repeated injections. Although a variety of SR drugs are already used in lab animal medicine, no studies exist that evaluate the suitability of an SR NSAID in sheep. This study used HPLC-MS to measure the plasma concentrations of 2 formulations of meloxicam-conventional and SRM- after subcutaneous administration in 6 adult ewes. Blood was collected at 0, 4, 12, 24, 36, 48, 60, 72, 84, 96, 120, 144, and 168 h after injection. In addition, physical exams, urinalysis, and biochemical analysis were performed at 0, 24, 48, and 120 h after dosage. Peak plasma concentrations were 1057 ± 433 ng/mL at 4 ± 0 h for conventional meloxicam and 3238 ± 1480 ng/mL at 6.7 ± 4.1 h for SR meloxicam (SRM). Elimination half-lives were 12.1 4.2 for CM and 15.2 ± 2.4 h for SRM. One sheep had an episode of acute renal azotemia starting 24 h after SRM administration; the episode resolved over time, and the definitive relationship to SRM administration was not determined. Plasma levels of SRM were higher than CM throughout the initial 24 h, remained variably elevated until 60 h after injection, but failed to sustain presumed therapeutic levels of 400 ng/mL for the full 72 h across all animals in this study. Further investigation is warranted to determine the safety and clinical efficacy of SRM in sheep. Currently, when SRM is used in sheep, we recommend the combination of a preemptive and multimodal analgesia regimen with clinical assessments throughout the postoperative period.
Collapse
Affiliation(s)
- Misha L Dunbar
- Research Animal Resources, Department of Surgery, University of Minnesota, Minneapolis, Minnesota;,
| | - Krista J Walkowiak
- Research Animal Resources, Department of Surgery, University of Minnesota, Minneapolis, Minnesota
| | - Jill Schappa Faustich
- Experimental Surgical Services, Department of Surgery, University of Minnesota, Minneapolis, Minnesota
| | - Aaron K Rendahl
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
| | - Melanie L Graham
- Preclinical Research Center, Department of Surgery, University of Minnesota, St Paul, Minnesota; Veterinary Population Medicine Department, University of Minnesota, St Paul, Minnesota
| |
Collapse
|
49
|
Musk GC, Kershaw H, Tano K, Niklasson A, von Unge M, Dilley RJ. Reactions to Gudair® vaccination identified in sheep used for biomedical research. Aust Vet J 2019; 97:56-60. [DOI: 10.1111/avj.12788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 10/26/2018] [Accepted: 01/15/2019] [Indexed: 12/29/2022]
Affiliation(s)
- GC Musk
- Animal Care Services; The University of Western Australia; M720, 35 Stirling Hwy, Crawley 6009 Western Australia Australia
| | - H Kershaw
- Animal Care Services; The University of Western Australia; M720, 35 Stirling Hwy, Crawley 6009 Western Australia Australia
| | - K Tano
- Department of Clinical Science, Otorhinolaryngology; Umeå University; Umeå Sweden
| | - A Niklasson
- Department of Clinical Science, Otorhinolaryngology; Umeå University; Umeå Sweden
| | - M von Unge
- Department of Clinical Medicine; Division of Surgery, University of Oslo; Oslo Norway
- Akershus University Hospital; Lorenskog Norway
| | - RJ Dilley
- Ear Sciences Centre; School of Medicine, the University of Western Australia; Crawley WA Australia
- Ear Science Institute Australia; Subiaco WA Australia
| |
Collapse
|
50
|
Saxena VK, Diaz A, Scheerlinck JPY. Identification and characterization of an M cell marker in nasopharynx- and oropharynx-associated lymphoid tissue of sheep. Vet Immunol Immunopathol 2019; 208:1-5. [DOI: 10.1016/j.vetimm.2018.12.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/12/2018] [Accepted: 12/17/2018] [Indexed: 01/10/2023]
|