1
|
Galili U. Accelerated Burn Healing in a Mouse Experimental Model Using α-Gal Nanoparticles. Bioengineering (Basel) 2023; 10:1165. [PMID: 37892895 PMCID: PMC10604883 DOI: 10.3390/bioengineering10101165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Macrophages play a pivotal role in the process of healing burns. One of the major risks in the course of burn healing, in the absence of regenerating epidermis, is infections, which greatly contribute to morbidity and mortality in such patients. Therefore, it is widely agreed that accelerating the recruitment of macrophages into burns may contribute to faster regeneration of the epidermis, thus decreasing the risk of infections. This review describes a unique method for the rapid recruitment of macrophages into burns and the activation of these macrophages to mediate accelerated regrowth of the epidermis and healing of burns. The method is based on the application of bio-degradable "α-gal" nanoparticles to burns. These nanoparticles present multiple α-gal epitopes (Galα1-3Galβ1-4GlcNAc-R), which bind the abundant natural anti-Gal antibody that constitutes ~1% of immunoglobulins in humans. Anti-Gal/α-gal nanoparticle interaction activates the complement system, resulting in localized production of the complement cleavage peptides C5a and C3a, which are highly effective chemotactic factors for monocyte-derived macrophages. The macrophages recruited into the α-gal nanoparticle-treated burns are activated following interaction between the Fc portion of anti-Gal coating the nanoparticles and the multiple Fc receptors on macrophage cell membranes. The activated macrophages secrete a variety of cytokines/growth factors that accelerate the regrowth of the epidermis and regeneration of the injured skin, thereby cutting the healing time by half. Studies on the healing of thermal injuries in the skin of anti-Gal-producing mice demonstrated a much faster recruitment of macrophages into burns treated with α-gal nanoparticles than in control burns treated with saline and healing of the burns within 6 days, whereas healing of control burns took ~12 days. α-Gal nanoparticles are non-toxic and do not cause chronic granulomas. These findings suggest that α-gal nanoparticles treatment may harness anti-Gal for inducing similar accelerated burn healing effects also in humans.
Collapse
Affiliation(s)
- Uri Galili
- Department of Medicine, Rush University Medical College, Chicago, IL 60612, USA
| |
Collapse
|
2
|
Chatanaka MK, Sohaei D, Diamandis EP, Prassas I. Beyond the amyloid hypothesis: how current research implicates autoimmunity in Alzheimer's disease pathogenesis. Crit Rev Clin Lab Sci 2023; 60:398-426. [PMID: 36941789 DOI: 10.1080/10408363.2023.2187342] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 03/01/2023] [Indexed: 03/23/2023]
Abstract
The amyloid hypothesis has so far been at the forefront of explaining the pathogenesis of Alzheimer's Disease (AD), a progressive neurodegenerative disorder that leads to cognitive decline and eventual death. Recent evidence, however, points to additional factors that contribute to the pathogenesis of this disease. These include the neurovascular hypothesis, the mitochondrial cascade hypothesis, the inflammatory hypothesis, the prion hypothesis, the mutational accumulation hypothesis, and the autoimmunity hypothesis. The purpose of this review was to briefly discuss the factors that are associated with autoimmunity in humans, including sex, the gut and lung microbiomes, age, genetics, and environmental factors. Subsequently, it was to examine the rise of autoimmune phenomena in AD, which can be instigated by a blood-brain barrier breakdown, pathogen infections, and dysfunction of the glymphatic system. Lastly, it was to discuss the various ways by which immune system dysregulation leads to AD, immunomodulating therapies, and future directions in the field of autoimmunity and neurodegeneration. A comprehensive account of the recent research done in the field was extracted from PubMed on 31 January 2022, with the keywords "Alzheimer's disease" and "autoantibodies" for the first search input, and "Alzheimer's disease" with "IgG" for the second. From the first search, 19 papers were selected, because they contained recent research on the autoantibodies found in the biofluids of patients with AD. From the second search, four papers were selected. The analysis of the literature has led to support the autoimmune hypothesis in AD. Autoantibodies were found in biofluids (serum/plasma, cerebrospinal fluid) of patients with AD with multiple methods, including ELISA, Mass Spectrometry, and microarray analysis. Through continuous research, the understanding of the synergistic effects of the various components that lead to AD will pave the way for better therapeutic methods and a deeper understanding of the disease.
Collapse
Affiliation(s)
- Miyo K Chatanaka
- Department of Laboratory and Medicine Pathobiology, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Dorsa Sohaei
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Eleftherios P Diamandis
- Department of Laboratory and Medicine Pathobiology, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
- Department of Clinical Biochemistry, University Health Network, Toronto, Canada
| | - Ioannis Prassas
- Laboratory Medicine Program, University Health Network, Toronto, Canada
| |
Collapse
|
3
|
Galili U. Antibody production and tolerance to the α-gal epitope as models for understanding and preventing the immune response to incompatible ABO carbohydrate antigens and for α-gal therapies. Front Mol Biosci 2023; 10:1209974. [PMID: 37449060 PMCID: PMC10338101 DOI: 10.3389/fmolb.2023.1209974] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023] Open
Abstract
This review describes the significance of the α-gal epitope (Galα-3Galβ1-4GlcNAc-R) as the core of human blood-group A and B antigens (A and B antigens), determines in mouse models the principles underlying the immune response to these antigens, and suggests future strategies for the induction of immune tolerance to incompatible A and B antigens in human allografts. Carbohydrate antigens, such as ABO antigens and the α-gal epitope, differ from protein antigens in that they do not interact with T cells, but B cells interacting with them require T-cell help for their activation. The α-gal epitope is the core of both A and B antigens and is the ligand of the natural anti-Gal antibody, which is abundant in all humans. In A and O individuals, anti-Gal clones (called anti-Gal/B) comprise >85% of the so-called anti-B activity and bind to the B antigen in facets that do not include fucose-linked α1-2 to the core α-gal. As many as 1% of B cells are anti-Gal B cells. Activation of quiescent anti-Gal B cells upon exposure to α-gal epitopes on xenografts and some protozoa can increase the titer of anti-Gal by 100-fold. α1,3-Galactosyltransferase knockout (GT-KO) mice lack α-gal epitopes and can produce anti-Gal. These mice simulate human recipients of ABO-incompatible human allografts. Exposure for 2-4 weeks of naïve and memory mouse anti-Gal B cells to α-gal epitopes in the heterotopically grafted wild-type (WT) mouse heart results in the elimination of these cells and immune tolerance to this epitope. Shorter exposures of 7 days of anti-Gal B cells to α-gal epitopes in the WT heart result in the production of accommodating anti-Gal antibodies that bind to α-gal epitopes but do not lyse cells or reject the graft. Tolerance to α-gal epitopes due to the elimination of naïve and memory anti-Gal B cells can be further induced by 2 weeks in vivo exposure to WT lymphocytes or autologous lymphocytes engineered to present α-gal epitopes by transduction of the α1,3-galactosyltransferase gene. These mouse studies suggest that autologous human lymphocytes similarly engineered to present the A or B antigen may induce corresponding tolerance in recipients of ABO-incompatible allografts. The review further summarizes experimental works demonstrating the efficacy of α-gal therapies in amplifying anti-viral and anti-tumor immune-protection and regeneration of injured tissues.
Collapse
Affiliation(s)
- Uri Galili
- Department of Medicine, Rush University Medical College, Chicago, IL, United States
| |
Collapse
|
4
|
Galili U. Paleo-immunology of human anti-carbohydrate antibodies preventing primate extinctions. Immunology 2023; 168:18-29. [PMID: 36161654 DOI: 10.1111/imm.13582] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/16/2022] [Indexed: 12/27/2022] Open
Abstract
Two human natural anti-carbohydrate antibodies appeared in critical evolutionary events that brought primates and hominins to brink of extinction. The first is the anti-Gal antibody, produced in Old-World monkeys (OWM), apes and humans. It binds the carbohydrate-antigen 'α-gal epitope' (Galα1-3Galβ1-4GlcNAc-R) on carbohydrate-chains (glycans) synthesized by non-primate mammals, lemurs and New-World monkeys (NWM). The second is anti-N-glycolylneuraminic-acid (anti-Neu5Gc) antibody binding Neu5Gc on glycans synthesized by OWM, apes and most non-primate mammals. Ancestral OWM and apes synthesized α-gal epitopes and were eliminated ~20-30 million-years-ago (mya). Only few accidentally mutated offspring lacking α-gal epitopes, produced anti-Gal and survived. Hominin-populations living ~3 mya synthesized Neu5Gc and were eliminated, but few mutated offspring that accidently lost their ability to synthesize Neu5Gc, produced natural anti-Neu5Gc antibody. These hominins survived and ultimately evolved into present-day humans. It is argued that these two near-extinction events were likely to be the result of epidemics caused by highly virulent and lethal enveloped viruses that killed parental-populations. These viruses presented α-gal epitopes or Neu5Gc synthesized in host-cells of the parental-populations. Mutated offspring survived the epidemics because they were protected from the lethal virus by the natural anti-Gal or anti-Neu5Gc antibodies they produced due to loss of immune-tolerance to α-gal epitopes or to Neu5Gc, respectively.
Collapse
Affiliation(s)
- Uri Galili
- Department of Medicine/Division of Cardiology, Rush University Medical College, Chicago, Illinois, USA
| |
Collapse
|
5
|
Galili U, Goldufsky JW, Schaer GL. α-Gal Nanoparticles Mediated Homing of Endogenous Stem Cells for Repair and Regeneration of External and Internal Injuries by Localized Complement Activation and Macrophage Recruitment. Int J Mol Sci 2022; 23:ijms231911490. [PMID: 36232789 PMCID: PMC9569695 DOI: 10.3390/ijms231911490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/27/2022] [Indexed: 12/02/2022] Open
Abstract
This review discusses a novel experimental approach for the regeneration of original tissue structure by recruitment of endogenous stem-cells to injured sites following administration of α-gal nanoparticles, which harness the natural anti-Gal antibody. Anti-Gal is produced in large amounts in all humans, and it binds the multiple α-gal epitopes (Galα1-3Galβ1-4GlcNAc-R) presented on α-gal nanoparticles. In situ binding of anti-Gal to α-gal nanoparticles activates the complement system and generates complement cleavage chemotactic-peptides that rapidly recruit macrophages. Macrophages reaching anti-Gal coated α-gal nanoparticles bind them via Fc/Fc receptor interaction and polarize into M2 pro-reparative macrophages. These macrophages secrete various cytokines that orchestrate regeneration of the injured tissue, including VEGF inducing neo-vascularization and cytokines directing homing of stem-cells to injury sites. Homing of stem-cells is also directed by interaction of complement cleavage peptides with their corresponding receptors on the stem-cells. Application of α-gal nanoparticles to skin wounds of anti-Gal producing mice results in decrease in healing time by half. Furthermore, α-gal nanoparticles treated wounds restore the normal structure of the injured skin without fibrosis or scar formation. Similarly, in a mouse model of occlusion/reperfusion myocardial-infarction, near complete regeneration after intramyocardial injection of α-gal nanoparticles was demonstrated, whereas hearts injected with saline display ~20% fibrosis and scar formation of the left ventricular wall. It is suggested that recruitment of stem-cells following anti-Gal/α-gal nanoparticles interaction in injured tissues may result in induction of localized regeneration facilitated by conducive microenvironments generated by pro-reparative macrophage secretions and “cues” provided by the extracellular matrix in the injury site.
Collapse
|
6
|
Galili U. Biosynthesis of α-Gal Epitopes (Galα1-3Galβ1-4GlcNAc-R) and Their Unique Potential in Future α-Gal Therapies. Front Mol Biosci 2021; 8:746883. [PMID: 34805272 PMCID: PMC8601398 DOI: 10.3389/fmolb.2021.746883] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/21/2021] [Indexed: 11/19/2022] Open
Abstract
The α-gal epitope is a carbohydrate antigen which appeared early in mammalian evolution and is synthesized in large amounts by the glycosylation enzyme α1,3galactosyltransferase (α1,3GT) in non-primate mammals, lemurs, and New-World monkeys. Ancestral Old-World monkeys and apes synthesizing α-gal epitopes underwent complete extinction 20–30 million years ago, and their mutated progeny lacking α-gal epitopes survived. Humans, apes, and Old-World monkeys which evolved from the surviving progeny lack α-gal epitopes and produce the natural anti-Gal antibody which binds specifically to α-gal epitopes. Because of this reciprocal distribution of the α-gal epitope and anti-Gal in mammals, transplantation of organs from non-primate mammals (e.g., pig xenografts) into Old-World monkeys or humans results in hyperacute rejection following anti-Gal binding to α-gal epitopes on xenograft cells. The in vivo immunocomplexing between anti-Gal and α-gal epitopes on molecules, pathogens, cells, or nanoparticles may be harnessed for development of novel immunotherapies (referred to as “α-gal therapies”) in various clinical settings because such immune complexes induce several beneficial immune processes. These immune processes include localized activation of the complement system which can destroy pathogens and generate chemotactic peptides that recruit antigen-presenting cells (APCs) such as macrophages and dendritic cells, targeting of antigens presenting α-gal epitopes for extensive uptake by APCs, and activation of recruited macrophages into pro-reparative macrophages. Some of the suggested α-gal therapies associated with these immune processes are as follows: 1. Increasing efficacy of enveloped-virus vaccines by synthesizing α-gal epitopes on vaccinating inactivated viruses, thereby targeting them for extensive uptake by APCs. 2. Conversion of autologous tumors into antitumor vaccines by expression of α-gal epitopes on tumor cell membranes. 3. Accelerating healing of external and internal injuries by α-gal nanoparticles which decrease the healing time and diminish scar formation. 4. Increasing anti-Gal–mediated protection against zoonotic viruses presenting α-gal epitopes and against protozoa, such as Trypanosoma, Leishmania, and Plasmodium, by vaccination for elevating production of the anti-Gal antibody. The efficacy and safety of these therapies were demonstrated in transgenic mice and pigs lacking α-gal epitopes and producing anti-Gal, raising the possibility that these α-gal therapies may be considered for further evaluation in clinical trials.
Collapse
Affiliation(s)
- Uri Galili
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
7
|
Kikuchi T, Nishimura M, Hirata M, Tanihara F, Komori N, Tanaka M, Sawamoto O, Otoi T, Matsumoto S. Development and characterization of Gal KO porcine bone marrow-derived mesenchymal stem cells. Xenotransplantation 2021; 28:e12717. [PMID: 34730861 DOI: 10.1111/xen.12717] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/27/2021] [Accepted: 10/11/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND We demonstrated that neonatal porcine bone marrow-derived mesenchymal stem cell (npBM-MSCs) could improve a critical ischemic limb disease in rat model more efficiently compared with human MSCs. However, since porcine MSC presents galactosyl-alpha 1,3-galactose antigen (Gal antigen), MSC could be eliminated by the xenogeneic rejection. Recently, we established Gal knockout (KO) pigs by a technique of the electroporation of the CRISPR/Cas9 system into vitro-fertilized zygotes. In this study, we hypothesized that MSC from the established Gal KO pigs could further improve the efficacy. Before examining the hypothesis, in this study, we have established and characterized bone marrow-derived MSC from the Gal KO adult pigs (apBM-MSCs). METHODS Mononuclear cells (MNCs) were isolated from bone marrow cells of both Gal KO adult pigs and wild-type (WT) adult pigs. MNCs were further manipulated to create Gal KO apBM-MSCs and WT apBM-MSCs. Both MSCs were assessed by their surface markers, the capability of differentiation into adipocytes, osteocytes and chondrocytes, grow speed and colony-forming assay. To assess the efficacy of Gal KO apBM-MSCs, angiogenesis-related genes and immunosuppression-related genes were assessed by cytokine stimulation. RESULTS Gal KO apBM-MSC showed no Gal antigen on their cell surfaces. Both Gal KO apBM-MSCs and WT apBM-MSCs, presented little or no negative surface markers of MSCs, while they presented positive surface markers of MSCs. Furthermore, Gal KO apBM-MSCs were able to differentiate into adipocytes, osteocytes, and chondrocytes as well as WT apBM-MSCs. There was no difference in doubling time between Gal KO apBM-MSCs and WT apBM-MSCs. Interestingly, the colony-forming efficiency of Gal KO apBM-MSCs was about half that of WT apBM-MSC. However, angiogenesis and immunosuppression-related genes were equally upregulated in both Gal KO apBM-MSCs and WT apBM-MSCs by cytokine stimulation. CONCLUSION We created and characterized Gal KO apBM-MSCs which showed similar characteristics and cytokine-induced gene upregulation to the WT apBM-MSCs.
Collapse
Affiliation(s)
- Takeshi Kikuchi
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc., Naruto, Tokushima, Japan
| | - Masuhiro Nishimura
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc., Naruto, Tokushima, Japan
| | - Maki Hirata
- Faculty of Bioscience and Bioindustry, Tokushima University, Myozai-gun, Tokushima, Japan
| | - Fuminori Tanihara
- Faculty of Bioscience and Bioindustry, Tokushima University, Myozai-gun, Tokushima, Japan
| | - Natsuki Komori
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc., Naruto, Tokushima, Japan
| | - Makoto Tanaka
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc., Naruto, Tokushima, Japan
| | - Osamu Sawamoto
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc., Naruto, Tokushima, Japan
| | - Takeshige Otoi
- Faculty of Bioscience and Bioindustry, Tokushima University, Myozai-gun, Tokushima, Japan
| | - Shinichi Matsumoto
- Research and Development Center, Otsuka Pharmaceutical Factory, Inc., Naruto, Tokushima, Japan
| |
Collapse
|
8
|
Ground M, Waqanivavalagi S, Walker R, Milsom P, Cornish J. Models of immunogenicity in preclinical assessment of tissue engineered heart valves. Acta Biomater 2021; 133:102-113. [PMID: 34082103 DOI: 10.1016/j.actbio.2021.05.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 12/20/2022]
Abstract
Tissue engineered heart valves may one day offer an exciting alternative to traditional valve prostheses. Methods of construction vary, from decellularised animal tissue to synthetic hydrogels, but the goal is the same: the creation of a 'living valve' populated with autologous cells that may persist indefinitely upon implantation. Previous failed attempts in humans have highlighted the difficulty in predicting how a novel heart valve will perform in vivo. A significant hurdle in bringing these prostheses to market is understanding the immune reaction in the short and long term. With respect to innate immunity, the chronic remodelling of a tissue engineered implant by macrophages remains poorly understood. Also unclear are the mechanisms behind unknown antigens and their effect on the adaptive immune system. No silver bullet exists, rather researchers must draw upon a number of in vitro and in vivo models to fully elucidate the effect a host will exert on the graft. This review details the methods by which the immunogenicity of tissue engineered heart valves may be investigated and reveals areas that would benefit from more research. STATEMENT OF SIGNIFICANCE: Both academic and private institutions around the world are committed to the creation of a valve prosthesis that will perform safely upon implantation. To date, however, no truly non-immunogenic valves have emerged. This review highlights the importance of preclinical immunogenicity assessment, and summarizes the available techniques used in vitro and in vivo to elucidate the immune response. To the authors knowledge, this is the first review that details the immune testing regimen specific to a TEHV candidate.
Collapse
|
9
|
Galili U, Zhu Z, Chen J, Goldufsky JW, Schaer GL. Near Complete Repair After Myocardial Infarction in Adult Mice by Altering the Inflammatory Response With Intramyocardial Injection of α-Gal Nanoparticles. Front Cardiovasc Med 2021; 8:719160. [PMID: 34513957 PMCID: PMC8425953 DOI: 10.3389/fcvm.2021.719160] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/30/2021] [Indexed: 12/27/2022] Open
Abstract
Background: Neonatal mice, but not older mice, can regenerate their hearts after myocardial-infarction (MI), a process mediated by pro-reparative macrophages. α-Gal nanoparticles applied to skin wounds in adult-mice bind the anti-Gal antibody, activate the complement cascade and generate complement chemotactic peptides that recruit pro-reparative macrophages which are further activated by these nanoparticles. The recruited macrophages decrease wound healing time by ~50%, restore the normal skin structure and prevent fibrosis and scar formation in mice. Objectives: The objective of this study is to determine if α-gal nanoparticles injected into the reperfused myocardium after MI in adult-mice can induce myocardial repair that restores normal structure, similar to that observed in skin injuries. Methods and Results: MI was induced by occluding the mid-portion of the left anterior descending (LAD) coronary artery for 30 min. Immediately following reperfusion, each mouse received two 10 μl injections of 100 μg α-gal nanoparticles in saline into the LAD territory (n = 20), or saline for controls (n = 10). Myocardial infarct size was measured by planimetry following Trichrome staining and macrophage recruitment by hematoxylin-eosin staining. Left ventricular (LV) function was measured by echocardiography. Control mice displayed peak macrophage infiltration at 4-days, whereas treated mice had a delayed peak macrophage infiltration at 7-days. At 28-days, control mice demonstrated large transmural infarcts with extensive scar formation and poor contractile function. In contrast, mice treated with α-gal nanoparticles demonstrated after 28-days a marked reduction in infarct size (~10-fold smaller), restoration of normal myocardium structure and contractile function. Conclusions: Intramyocardial injection of α-gal nanoparticles post-MI in anti-Gal producing adult-mice results in near complete repair of the infarcted territory, with restoration of normal LV structure and contractile function. The mechanism responsible for this benefit likely involves alteration of the usual inflammatory response post-MI, as previously observed with regeneration of injured hearts in adult zebrafish, salamanders and neonatal mice.
Collapse
Affiliation(s)
- Uri Galili
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Zhongkai Zhu
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Jiwang Chen
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Josef W Goldufsky
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Gary L Schaer
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
10
|
In Situ "Humanization" of Porcine Bioprostheses: Demonstration of Tendon Bioprostheses Conversion into Human ACL and Possible Implications for Heart Valve Bioprostheses. Bioengineering (Basel) 2021; 8:bioengineering8010010. [PMID: 33445522 PMCID: PMC7826727 DOI: 10.3390/bioengineering8010010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 11/21/2022] Open
Abstract
This review describes the first studies on successful conversion of porcine soft-tissue bioprostheses into viable permanently functional tissue in humans. This process includes gradual degradation of the porcine tissue, with concomitant neo-vascularization and reconstruction of the implanted bioprosthesis with human cells and extracellular matrix. Such a reconstruction process is referred to in this review as “humanization”. Humanization was achieved with porcine bone-patellar-tendon-bone (BTB), replacing torn anterior-cruciate-ligament (ACL) in patients. In addition to its possible use in orthopedic surgery, it is suggested that this humanization method should be studied as a possible mechanism for converting implanted porcine bioprosthetic heart-valves (BHV) into viable tissue valves in young patients. Presently, these patients are only implanted with mechanical heart-valves, which require constant anticoagulation therapy. The processing of porcine bioprostheses, which enables humanization, includes elimination of α-gal epitopes and partial (incomplete) crosslinking with glutaraldehyde. Studies on implantation of porcine BTB bioprostheses indicated that enzymatic elimination of α-gal epitopes prevents subsequent accelerated destruction of implanted tissues by the natural anti-Gal antibody, whereas the partial crosslinking by glutaraldehyde molecules results in their function as “speed bumps” that slow the infiltration of macrophages. Anti-non gal antibodies produced against porcine antigens in implanted bioprostheses recruit macrophages, which infiltrate at a pace that enables slow degradation of the porcine tissue, neo-vascularization, and infiltration of fibroblasts. These fibroblasts align with the porcine collagen-fibers scaffold, secrete their collagen-fibers and other extracellular-matrix (ECM) components, and gradually replace porcine tissues degraded by macrophages with autologous functional viable tissue. Porcine BTB implanted in patients completes humanization into autologous ACL within ~2 years. The similarities in cells and ECM comprising heart-valves and tendons, raises the possibility that porcine BHV undergoing a similar processing, may also undergo humanization, resulting in formation of an autologous, viable, permanently functional, non-calcifying heart-valves.
Collapse
|
11
|
Mallis P, Kostakis A, Stavropoulos-Giokas C, Michalopoulos E. Future Perspectives in Small-Diameter Vascular Graft Engineering. Bioengineering (Basel) 2020; 7:E160. [PMID: 33321830 PMCID: PMC7763104 DOI: 10.3390/bioengineering7040160] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/04/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
The increased demands of small-diameter vascular grafts (SDVGs) globally has forced the scientific society to explore alternative strategies utilizing the tissue engineering approaches. Cardiovascular disease (CVD) comprises one of the most lethal groups of non-communicable disorders worldwide. It has been estimated that in Europe, the healthcare cost for the administration of CVD is more than 169 billion €. Common manifestations involve the narrowing or occlusion of blood vessels. The replacement of damaged vessels with autologous grafts represents one of the applied therapeutic approaches in CVD. However, significant drawbacks are accompanying the above procedure; therefore, the exploration of alternative vessel sources must be performed. Engineered SDVGs can be produced through the utilization of non-degradable/degradable and naturally derived materials. Decellularized vessels represent also an alternative valuable source for the development of SDVGs. In this review, a great number of SDVG engineering approaches will be highlighted. Importantly, the state-of-the-art methodologies, which are currently employed, will be comprehensively presented. A discussion summarizing the key marks and the future perspectives of SDVG engineering will be included in this review. Taking into consideration the increased number of patients with CVD, SDVG engineering may assist significantly in cardiovascular reconstructive surgery and, therefore, the overall improvement of patients' life.
Collapse
Affiliation(s)
- Panagiotis Mallis
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (C.S.-G.); (E.M.)
| | - Alkiviadis Kostakis
- Center of Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece;
| | - Catherine Stavropoulos-Giokas
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (C.S.-G.); (E.M.)
| | - Efstathios Michalopoulos
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (C.S.-G.); (E.M.)
| |
Collapse
|
12
|
Galili U. Amplifying immunogenicity of prospective Covid-19 vaccines by glycoengineering the coronavirus glycan-shield to present α-gal epitopes. Vaccine 2020; 38:6487-6499. [PMID: 32907757 PMCID: PMC7437500 DOI: 10.1016/j.vaccine.2020.08.032] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/22/2020] [Accepted: 08/12/2020] [Indexed: 12/16/2022]
Abstract
The many carbohydrate chains on Covid-19 coronavirus SARS-CoV-2 and its S-protein form a glycan-shield that masks antigenic peptides and decreases uptake of inactivated virus or S-protein vaccines by APC. Studies on inactivated influenza virus and recombinant gp120 of HIV vaccines indicate that glycoengineering of glycan-shields to present α-gal epitopes (Galα1-3Galβ1-4GlcNAc-R) enables harnessing of the natural anti-Gal antibody for amplifying vaccine efficacy, as evaluated in mice producing anti-Gal. The α-gal epitope is the ligand for the natural anti-Gal antibody which constitutes ~1% of immunoglobulins in humans. Upon administration of vaccines presenting α-gal epitopes, anti-Gal binds to these epitopes at the vaccination site and forms immune complexes with the vaccines. These immune complexes are targeted for extensive uptake by APC as a result of binding of the Fc portion of immunocomplexed anti-Gal to Fc receptors on APC. This anti-Gal mediated effective uptake of vaccines by APC results in 10-200-fold higher anti-viral immune response and in 8-fold higher survival rate following challenge with a lethal dose of live influenza virus, than same vaccines lacking α-gal epitopes. It is suggested that glycoengineering of carbohydrate chains on the glycan-shield of inactivated SARS-CoV-2 or on S-protein vaccines, for presenting α-gal epitopes, will have similar amplifying effects on vaccine efficacy. α-Gal epitope synthesis on coronavirus vaccines can be achieved with recombinant α1,3galactosyltransferase, replication of the virus in cells with high α1,3galactosyltransferase activity as a result of stable transfection of cells with several copies of the α1,3galactosyltransferase gene (GGTA1), or by transduction of host cells with replication defective adenovirus containing this gene. In addition, recombinant S-protein presenting multiple α-gal epitopes on the glycan-shield may be produced in glycoengineered yeast or bacteria expression systems containing the corresponding glycosyltransferases. Prospective Covid-19 vaccines presenting α-gal epitopes may provide better protection than vaccines lacking this epitope because of increased uptake by APC.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/biosynthesis
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Antigens, Viral/metabolism
- Betacoronavirus/drug effects
- Betacoronavirus/immunology
- Betacoronavirus/pathogenicity
- COVID-19
- COVID-19 Vaccines
- Coronavirus Infections/genetics
- Coronavirus Infections/immunology
- Coronavirus Infections/prevention & control
- Coronavirus Infections/virology
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/virology
- Genetic Engineering
- HIV Core Protein p24/chemistry
- HIV Core Protein p24/genetics
- HIV Core Protein p24/immunology
- HIV Envelope Protein gp120/chemistry
- HIV Envelope Protein gp120/genetics
- HIV Envelope Protein gp120/immunology
- Humans
- Immunogenicity, Vaccine
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/virology
- Mice
- Pandemics/prevention & control
- Pneumonia, Viral/immunology
- Pneumonia, Viral/prevention & control
- Pneumonia, Viral/virology
- Recombinant Fusion Proteins/chemistry
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- SARS-CoV-2
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/metabolism
- Trisaccharides/chemistry
- Trisaccharides/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/biosynthesis
- Viral Vaccines/genetics
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Uri Galili
- Department of Medicine, Rush Medical School, Chicago, IL, USA.
| |
Collapse
|
13
|
Host Synthesized Carbohydrate Antigens on Viral Glycoproteins as "Achilles' Heel" of Viruses Contributing to Anti-Viral Immune Protection. Int J Mol Sci 2020; 21:ijms21186702. [PMID: 32933166 PMCID: PMC7555091 DOI: 10.3390/ijms21186702] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/09/2020] [Accepted: 09/09/2020] [Indexed: 01/10/2023] Open
Abstract
The glycans on enveloped viruses are synthesized by host-cell machinery. Some of these glycans on zoonotic viruses of mammalian reservoirs are recognized by human natural antibodies that may protect against such viruses. These antibodies are produced mostly against carbohydrate antigens on gastrointestinal bacteria and fortuitously, they bind to carbohydrate antigens synthesized in other mammals, neutralize and destroy viruses presenting these antigens. Two such antibodies are: anti-Gal binding to α-gal epitopes synthesized in non-primate mammals, lemurs, and New World monkeys, and anti-N-glycolyl neuraminic acid (anti-Neu5Gc) binding to N-glycolyl-neuraminic acid (Neu5Gc) synthesized in apes, Old World monkeys, and many non-primate mammals. Anti-Gal appeared in Old World primates following accidental inactivation of the α1,3galactosyltransferase gene 20–30 million years ago. Anti-Neu5Gc appeared in hominins following the inactivation of the cytidine-monophosphate-N-acetyl-neuraminic acid hydroxylase gene, which led to the loss of Neu5Gc <6 million-years-ago. It is suggested that an epidemic of a lethal virus eliminated ancestral Old World-primates synthesizing α-gal epitopes, whereas few mutated offspring lacking α-gal epitopes and producing anti-Gal survived because anti-Gal destroyed viruses presenting α-gal epitopes, following replication in parental populations. Similarly, anti-Neu5Gc protected few mutated hominins lacking Neu5Gc in lethal virus epidemics that eliminated parental hominins synthesizing Neu5Gc. Since α-gal epitopes are presented on many zoonotic viruses it is suggested that vaccines elevating anti-Gal titers may be of protective significance in areas endemic for such zoonotic viruses. This protection would be during the non-primate mammal to human virus transmission, but not in subsequent human to human transmission where the virus presents human glycans. In addition, production of viral vaccines presenting multiple α-gal epitopes increases their immunogenicity because of effective anti-Gal-mediated targeting of vaccines to antigen presenting cells for extensive uptake of the vaccine by these cells.
Collapse
|
14
|
Galili U. Human Natural Antibodies to Mammalian Carbohydrate Antigens as Unsung Heroes Protecting against Past, Present, and Future Viral Infections. Antibodies (Basel) 2020; 9:E25. [PMID: 32580274 PMCID: PMC7344964 DOI: 10.3390/antib9020025] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 06/18/2020] [Indexed: 12/22/2022] Open
Abstract
Human natural antibodies to mammalian carbohydrate antigens (MCA) bind to carbohydrate-antigens synthesized in other mammalian species and protect against zoonotic virus infections. Three such anti-MCA antibodies are: (1) anti-Gal, also produced in Old-World monkeys and apes, binds to α-gal epitopes synthesized in non-primate mammals, lemurs, and New-World monkeys; (2) anti-Neu5Gc binds to Neu5Gc (N-glycolyl-neuraminic acid) synthesized in apes, Old-World monkeys, and many non-primate mammals; and (3) anti-Forssman binds to Forssman-antigen synthesized in various mammals. Anti-viral protection by anti-MCA antibodies is feasible because carbohydrate chains of virus envelopes are synthesized by host glycosylation machinery and thus are similar to those of their mammalian hosts. Analysis of MCA glycosyltransferase genes suggests that anti-Gal appeared in ancestral Old-World primates following catastrophic selection processes in which parental populations synthesizing α-gal epitopes were eliminated in enveloped virus epidemics. However, few mutated offspring in which the α1,3galactosyltransferase gene was accidentally inactivated produced natural anti-Gal that destroyed viruses presenting α-gal epitopes, thereby preventing extinction of mutated offspring. Similarly, few mutated hominin offspring that ceased to synthesize Neu5Gc produced anti-Neu5Gc, which destroyed viruses presenting Neu5Gc synthesized in parental hominin populations. A present-day example for few humans having mutations that prevent synthesis of a common carbohydrate antigen (produced in >99.99% of humans) is blood-group Bombay individuals with mutations inactivating H-transferase; thus, they cannot synthesize blood-group O (H-antigen) but produce anti-H antibody. Anti-MCA antibodies prevented past extinctions mediated by enveloped virus epidemics, presently protect against zoonotic-viruses, and may protect in future epidemics. Travelers to regions with endemic zoonotic viruses may benefit from vaccinations elevating protective anti-MCA antibody titers.
Collapse
Affiliation(s)
- Uri Galili
- Department of Medicine, Rush University Medical School, Chicago, IL 60605, USA
| |
Collapse
|
15
|
Tector AJ, Mosser M, Tector M, Bach JM. The Possible Role of Anti-Neu5Gc as an Obstacle in Xenotransplantation. Front Immunol 2020; 11:622. [PMID: 32351506 PMCID: PMC7174778 DOI: 10.3389/fimmu.2020.00622] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 03/18/2020] [Indexed: 12/20/2022] Open
Abstract
Seventy to ninety percentage of preformed xenoreactive antibodies in human serum bind to the galactose-α(1,3)-galactose Gal epitope, and the creation of Gal knockout (KO) pigs has eliminated hyperacute rejection as a barrier to xenotransplantation. Now other glycan antigens are barriers to move ahead with xenotransplantation, and the N-glycolyl neuraminic acid, Neu5Gc (or Hanganutziu-Deicher antigen), is also a major pig xenoantigen. Humans have anti-Neu5Gc antibodies. Several data indicate a strong immunogenicity of Neu5Gc in humans that may contribute to an important part in antibody-dependent injury to pig xenografts. Pig islets express Neu5Gc, which reacted with diet-derived human antibodies and mice deleted for Neu5Gc reject pancreatic islets from wild-type counterpart. However, Neu5Gc positive heart were not rejected in Neu5Gc KO mice indicating that the role of Neu5Gc-specific antibodies has to be nuanced and depend of the graft situation parameters (organ/tissue, recipient, implication of other glycan antigens). Recently generated Gal/Neu5Gc KO pigs eliminate the expression of Gal and Neu5Gc, and improve the crossmatch of humans with the pig. This review summarizes the current and recent experimental and (pre)clinical data on the Neu5Gc immunogenicity and emphasize of the potential impact of anti-Neu5Gc antibodies in limiting xenotransplantation in humans.
Collapse
Affiliation(s)
- Alfred Joseph Tector
- Department of Surgery, University of Alabama at Birmingham School of Medicine, Birmingham, AL, United States
| | - Mathilde Mosser
- Immuno-Endocrinology Unit (IECM), USC1383, Oniris, INRA, Nantes, France
| | - Matthew Tector
- Department of Surgery, University of Alabama at Birmingham School of Medicine, Birmingham, AL, United States
| | - Jean-Marie Bach
- Immuno-Endocrinology Unit (IECM), USC1383, Oniris, INRA, Nantes, France
| |
Collapse
|
16
|
Galili U. Evolution in primates by “Catastrophic‐selection” interplay between enveloped virus epidemics, mutated genes of enzymes synthesizing carbohydrate antigens, and natural anti‐carbohydrate antibodies. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2018; 168:352-363. [DOI: 10.1002/ajpa.23745] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/09/2018] [Accepted: 10/16/2018] [Indexed: 02/01/2023]
Affiliation(s)
- Uri Galili
- Department of MedicineRush Medical College Chicago Illinois
| |
Collapse
|
17
|
Kurome M, Baehr A, Simmet K, Jemiller EM, Egerer S, Dahlhoff M, Zakhartchenko V, Nagashima H, Klymiuk N, Kessler B, Wolf E. Targeting αGal epitopes for multi-species embryo immunosurgery. Reprod Fertil Dev 2018; 31:820-826. [PMID: 30384878 DOI: 10.1071/rd18120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 09/20/2018] [Indexed: 11/23/2022] Open
Abstract
Immunosurgical isolation of the inner cell mass (ICM) from blastocysts is based on complement-mediated lysis of antibody-coated trophectoderm (TE) cells. Conventionally, anti-species antisera, containing antibodies against multiple undefined TE-cell epitopes, have been used as the antibody source. We previously generated α-1,3-galactosyltransferase deficient (GTKO) pigs to prevent hyperacute rejection of pig-to-primate xenotransplants. Since GTKO pigs lack galactosyl-α-1,3-galactose (αGal) but are exposed to this antigen (e.g. αGal on gut bacteria), they produce anti-αGal antibodies. In this study, we examined whether serum from GTKO pigs could be used as a novel antibody source for multi-species embryo immunosurgery. Mouse, rabbit, pig and cattle blastocysts were used for the experiment. Expression of αGal epitopes on the surface of TE cells was detected in blastocysts of all species tested. GTKO pig serum contained sufficient anti-αGal antibodies to induce complement-mediated lysis of TE cells in blastocysts from all species investigated. Intact ICMs could be successfully recovered and the majority showed the desired level of purity. Our study demonstrates that GTKO pig serum is a reliable and effective source of antibodies targeting the αGal epitopes of TE cells for multi-species embryo immunosurgery.
Collapse
Affiliation(s)
- Mayuko Kurome
- Chair for Molecular Animal Breeding and Biotechnology, Centre for Innovative Medical Models (CiMM), LMU Munich, Hacker strasse 27, 85764 Oberschleissheim, Germany
| | - Andrea Baehr
- Klinikum Rechts der Isar, Innere Medizin I, TU Munich, Ismaninger strasse 22, 81675 Munich, Germany
| | - Kilian Simmet
- Chair for Molecular Animal Breeding and Biotechnology, Centre for Innovative Medical Models (CiMM), LMU Munich, Hacker strasse 27, 85764 Oberschleissheim, Germany
| | - Eva-Maria Jemiller
- Chair for Molecular Animal Breeding and Biotechnology, Centre for Innovative Medical Models (CiMM), LMU Munich, Hacker strasse 27, 85764 Oberschleissheim, Germany
| | - Stefanie Egerer
- Chair for Molecular Animal Breeding and Biotechnology, Centre for Innovative Medical Models (CiMM), LMU Munich, Hacker strasse 27, 85764 Oberschleissheim, Germany
| | - Maik Dahlhoff
- Chair for Molecular Animal Breeding and Biotechnology, Centre for Innovative Medical Models (CiMM), LMU Munich, Hacker strasse 27, 85764 Oberschleissheim, Germany
| | - Valeri Zakhartchenko
- Chair for Molecular Animal Breeding and Biotechnology, Centre for Innovative Medical Models (CiMM), LMU Munich, Hacker strasse 27, 85764 Oberschleissheim, Germany
| | - Hiroshi Nagashima
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama, Kawasaki, Kanagawa 214-8571, Japan
| | - Nikolai Klymiuk
- Chair for Molecular Animal Breeding and Biotechnology, Centre for Innovative Medical Models (CiMM), LMU Munich, Hacker strasse 27, 85764 Oberschleissheim, Germany
| | - Barbara Kessler
- Chair for Molecular Animal Breeding and Biotechnology, Centre for Innovative Medical Models (CiMM), LMU Munich, Hacker strasse 27, 85764 Oberschleissheim, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Centre for Innovative Medical Models (CiMM), LMU Munich, Hacker strasse 27, 85764 Oberschleissheim, Germany
| |
Collapse
|
18
|
Lu Y, Shao A, Shan Y, Zhao H, Leiguo M, Zhang Y, Tang Y, Zhang W, Jin Y, Xu L. A standardized quantitative method for detecting remnant alpha-Gal antigen in animal tissues or animal tissue-derived biomaterials and its application. Sci Rep 2018; 8:15424. [PMID: 30337555 PMCID: PMC6194003 DOI: 10.1038/s41598-018-32959-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/27/2018] [Indexed: 01/19/2023] Open
Abstract
Alpha-Gal (Gal) epitopes present in animal tissues are known to be the key xenoantigens that elicit xenorejection. However, a standardized method to determine Gal epitope in animal tissue-derived biomaterials does not exist. Herein, a standardized method for quantitative detection of Gal antigen was established based on an ELISA inhibition assay with Gal antibody. In this method, the key optimized experimental conditions were: (1) Gal-antigen positive and negative reference materials were developed, and used as positive and negative control in the test system, respectively; (2) A mixture of artificial Gal-BSA antigen plus Gal-negative matrix was used as the calibration standard sample, making it has similar composition with test sample; and (3) The lysis buffer was combined with the homogenate to expose the Gal antigen as much as possible. The results from validation and application experiments showed that the standardized method had good reproducibility (RSD = 12.48%), and the lower detection limit (LDL) is ~7.1 × 1011 Gal epitopes/reaction. This method has been further developed into a detection Kit (Meitan 70101, China), and it has been developed as a standard method for detecting remnant immunogen of animal tissue derived medical devices, and as the industry standard has been released in China. (YY/T 1561–2017).
Collapse
Affiliation(s)
- Yan Lu
- National Institutes for Food and Drug Control, 102629, Beijing, China.,School of Medical Lab Science and life Science, Wenzhou Medical University, 325035, Wenzhou, China.,Subei People's Hospital of Jiangsu Province, 225001, Jiangsu, China
| | - Anliang Shao
- National Institutes for Food and Drug Control, 102629, Beijing, China
| | - Yongqiang Shan
- National Institutes for Food and Drug Control, 102629, Beijing, China.,School of Medical Lab Science and life Science, Wenzhou Medical University, 325035, Wenzhou, China
| | - Hongni Zhao
- Research and Development Center for Tissue Engineering, Fourth Military Medical University, 710032, Xi'an, China
| | - Ming Leiguo
- Research and Development Center for Tissue Engineering, Fourth Military Medical University, 710032, Xi'an, China
| | - Yongjie Zhang
- Research and Development Center for Tissue Engineering, Fourth Military Medical University, 710032, Xi'an, China
| | - Yinxi Tang
- National Engineering Laboratory for Regenerative Medical Implant Devices, Guanhao Biotech, Co., LTD, 510530, Guangzhou, China
| | - Wei Zhang
- National Engineering Laboratory for Regenerative Medical Implant Devices, Guanhao Biotech, Co., LTD, 510530, Guangzhou, China
| | - Yan Jin
- Research and Development Center for Tissue Engineering, Fourth Military Medical University, 710032, Xi'an, China.
| | - Liming Xu
- National Institutes for Food and Drug Control, 102629, Beijing, China. .,School of Medical Lab Science and life Science, Wenzhou Medical University, 325035, Wenzhou, China.
| |
Collapse
|
19
|
Analysis of Leishmania mimetic neoglycoproteins for the cutaneous leishmaniasis diagnosis. Parasitology 2018; 145:1938-1948. [PMID: 29806570 DOI: 10.1017/s0031182018000720] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Oligosaccharides are broadly present on Leishmania cell surfaces. They can be useful for the leishmaniases diagnosis and also helpful in identifying new cell markers for the disease. The disaccharide Galα1-3Galβ is the immunodominant saccharide in Leishmania cell surface and is the unique non-reducing terminal glycosphingolipids structure recognized by anti-α-Gal. This study describes an enzyme-linked immunosorbent assay (ELISA) used to measure serum levels of anti-α-galactosyl (α-Gal) antibodies in patients with cutaneous leishmaniasis (CL). Optimal ELISA conditions were established and two neoglycoproteins (NGP) containing the Galα1-3Gal terminal fraction (Galα1-3Galβ1-4GlcNAc-HAS and Galα1-3Gal-HAS) and one Galα1-3Gal NGP analogue (Galα1-3Galβ1-3GlcNAc-HAS) were used as antigens. Means of anti-α-Gal antibody titres of CL patients were significantly higher (P < 0.05) than the healthy individuals for all NGPs tested. Sensitivity and specificity of all NGPs ranged from 62.2 to 78.4% and 58.3 to 96.7%, respectively. In conclusion, the NGPs can be used for CL diagnosis.
Collapse
|
20
|
Abstract
The natural anti-Gal antibody is one of the multiple natural anti-carbohydrate antibodies produced in humans against a wide range of carbohydrate antigens on GI bacteria. The antibody is unique to humans, apes, and Old World monkeys, and it binds specifically to a mammalian carbohydrate antigen called the α-gal epitope that is synthesized in nonprimate mammals, lemurs (prosimians) and New World monkeys by the glycosylation enzyme α1,3GT. The α1,3GT gene (GGTA1) appeared in mammals >100 million years ago, prior to the split between marsupial and placental mammals. This gene has been conserved in its active form, in all mammals, except for Old World monkeys, apes, and humans. Inactivation of the α1,3GT gene in ancestral Old World primates occurred 20–30 million years ago and could have been associated with epidemics of enveloped viruses in the Eurasia-Africa continent. It is suggested that prior to such epidemics, few ancestral Old World primates acquired deletion point mutations that inactivated the α1,3GT gene and eliminated α-gal epitopes. This resulted in loss of immune tolerance to the α-gal epitope and thus, in production of the anti-Gal antibody against antigens on bacteria colonizing the GI tract. This accidental inactivation of the α1,3GT gene in very small populations is analogous to the highly rare blood type “Bombay” individuals who do not synthesize blood group H (O antigen) because of inactivation of the α1,2-fucosyltransferase gene. The loss of immune tolerance to blood group H antigen has resulted in production of natural anti-blood group H antibodies in the blood group Bombay individuals. It is suggested that anti-Gal protected against infections by enveloped viruses presenting α-gal epitopes, which were lethal to the parental primate populations that conserved active α1,3GT and thus, synthesized α-gal epitopes. Alternative causes for the elimination of Old World primates synthesizing α-gal epitopes could be bacteria or protozoa parasites presenting α-gal or α-gal-like epitopes, and bacterial toxins, or detrimental viruses that used α-gal epitopes in these primates as “docking receptors.” Ultimately, any of these proposed selective processes could result in extinction of Old World primates synthesizing α-gal epitopes on their cells. These ancestral primates were replaced by offspring populations lacking α-gal epitopes and producing the anti-Gal antibody, which continues to be produced by Old World monkeys, apes, and humans. New World monkeys and lemurs were protected from pathogens of the Old World by oceanic barriers, thus they continue to synthesize α-gal epitopes and lack the ability to produce the anti-Gal antibody. This scenario of few individuals in a large population having a mutation(s) that inactivates a glycosyltransferase gene thus, resulting in production of evolutionary advantageous natural antibodies against the eliminated carbohydrate antigen, may reflect one of the mechanisms inducing changes in the carbohydrate profile of various mammalian populations.
Collapse
|
21
|
LEA29Y expression in transgenic neonatal porcine islet-like cluster promotes long-lasting xenograft survival in humanized mice without immunosuppressive therapy. Sci Rep 2017; 7:3572. [PMID: 28620237 PMCID: PMC5472587 DOI: 10.1038/s41598-017-03913-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/05/2017] [Indexed: 02/02/2023] Open
Abstract
Genetically engineered pigs are a promising source for islet cell transplantation in type 1 diabetes, but the strong human anti-pig immune response prevents its successful clinical application. Here we studied the efficacy of neonatal porcine islet-like cell clusters (NPICCs) overexpressing LEA29Y, a high-affinity variant of the T cell co-stimulation inhibitor CTLA-4Ig, to engraft and restore normoglycemia after transplantation into streptozotocin-diabetic NOD-SCID IL2rγ−/− (NSG) mice stably reconstituted with a human immune system. Transplantation of INSLEA29Y expressing NPICCs resulted in development of normal glucose tolerance (70.4%) and long-term maintenance of normoglycemia without administration of immunosuppressive drugs. All animals transplanted with wild-type NPICCs remained diabetic. Immunohistological examinations revealed a strong peri- and intragraft infiltration of wild-type NPICCs with human CD45+ immune cells consisting of predominantly CD4+ and CD8+ lymphocytes and some CD68+ macrophages and FoxP3+ regulatory T cells. Significantly less infiltrating lymphocytes and only few macrophages were observed in animals transplanted with INSLEA29Y transgenic NPICCs. This is the first study providing evidence that beta cell-specific LEA29Y expression is effective for NPICC engraftment in the presence of a humanized immune system and it has a long-lasting protective effect on inhibition of human anti-pig xenoimmunity. Our findings may have important implications for the development of a low-toxic protocol for porcine islet transplantation in patients with type 1 diabetes.
Collapse
|
22
|
Galili U. α-Gal Nanoparticles in Wound and Burn Healing Acceleration. Adv Wound Care (New Rochelle) 2017; 6:81-92. [PMID: 28289553 PMCID: PMC5346952 DOI: 10.1089/wound.2016.0703] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 10/02/2016] [Indexed: 12/26/2022] Open
Abstract
Significance: Rapid recruitment and activation of macrophages may accelerate wound healing. Such accelerated healing was observed in wounds and burns of experimental animals treated with α-gal nanoparticles. Recent Advances: α-Gal nanoparticles present multiple α-gal epitopes (Galα1-3Galβ1-4GlcNAc-R). α-Gal nanoparticles applied to wounds bind anti-Gal (the most abundant antibody in humans) and generate chemotactic complement peptides, which rapidly recruit macrophages. Fc/Fc receptor interaction between anti-Gal coating the α-gal nanoparticles and recruited macrophages activates macrophages to produce cytokines that accelerate healing. α-Gal nanoparticles applied to burns and wounds in mice and pigs producing anti-Gal, decreased healing time by 40-60%. In mice, this accelerated healing avoided scar formation. α-Gal nanoparticle-treated wounds, in diabetic mice producing anti-Gal, healed within 12 days, whereas saline-treated wounds became chronic wounds. α-Gal nanoparticles are stable for years and may be applied dried, in suspension, aerosol, ointments, or within biodegradable materials. Critical Issues: α-Gal nanoparticle therapy can be evaluated only in mammalian models producing anti-Gal, including α1,3-galactosyltransferase knockout mice and pigs or Old World primates. Traditional experimental animal models synthesize α-gal epitopes and lack anti-Gal. Future Directions: Since anti-Gal is naturally produced in all humans, it is of interest to determine safety and efficacy of α-gal nanoparticles in accelerating wound and burn healing in healthy individuals and in patients with impaired wound healing such as diabetic patients and elderly individuals. In addition, efficacy of α-gal nanoparticle therapy should be studied in healing and regeneration of internal injuries such as surgical incisions, ischemic myocardium following myocardial infarction, and injured nerves.
Collapse
Affiliation(s)
- Uri Galili
- Correspondence: 910 South Michigan Avenue, Apartment 1404, Chicago, IL 60605(e-mail: )
| |
Collapse
|
23
|
Jones G, Herbert A, Berry H, Edwards JH, Fisher J, Ingham E. Decellularization and Characterization of Porcine Superflexor Tendon: A Potential Anterior Cruciate Ligament Replacement. Tissue Eng Part A 2017; 23:124-134. [PMID: 27806678 PMCID: PMC5312612 DOI: 10.1089/ten.tea.2016.0114] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The porcine superflexor tendon (SFT) was identified as having appropriate structure and properties for development of a decellularized device for use in anterior cruciate ligament reconstruction. SFTs were decellularized using a combination of freeze-thaw and washes in hypotonic buffer and 0.1% (w/v) sodium dodecyl sulfate in hypotonic buffer plus proteinase inhibitors, followed by nuclease treatment and sterilization using peracetic acid. The decellularized biological scaffold was devoid of cells and cell remnants and contained only 13 ng/mg (dry weight) residual total DNA. Immunohistochemistry showed retention of collagen type I and III and tenascin-C. Quantitative analysis of sulfated sugar and hydroxyproline content revealed a loss of glycosaminoglycans compared with native tissue, but no loss of collagen. The decellularized SFT was biocompatible in vitro and in vivo following implantation in a mouse subcutaneous model for 12 weeks. Uniaxial tensile testing to failure indicated that the gross material properties of decellularized SFTs were not significantly different to native tissue. Decellularized SFTs had an ultimate tensile strength of 61.8 ± 10.3 MPa (±95% confidence limits), a failure strain of 0.29 ± 0.04, and a Young's modulus of the collagen phase of 294.1 ± 61.9 MPa. Analysis of the presence of the α-Gal (galactose-α-1,3-galactose) epitope by immunohistochemistry, lectin binding, and antibody absorption assay indicated that the epitope was reduced, but still present post decellularization. This is discussed in light of the potential role of noncellular α-Gal in the acceleration of wound healing and tissue regeneration in the presence of antibodies to α-Gal.
Collapse
Affiliation(s)
- Gemma Jones
- Institute of Medical and Biological Engineering, School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Anthony Herbert
- Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, Leeds, United Kingdom
| | - Helen Berry
- Tissue Regenix Group, The Biocentre Innovation Way, York, United Kingdom
| | - Jennifer Helen Edwards
- Institute of Medical and Biological Engineering, School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - John Fisher
- Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, Leeds, United Kingdom
| | - Eileen Ingham
- Institute of Medical and Biological Engineering, School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
24
|
Galili U. Natural anti-carbohydrate antibodies contributing to evolutionary survival of primates in viral epidemics? Glycobiology 2016; 26:1140-1150. [PMID: 27567275 DOI: 10.1093/glycob/cww088] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 08/09/2016] [Accepted: 08/16/2016] [Indexed: 12/17/2022] Open
Abstract
Humans produce multiple natural antibodies against carbohydrate antigens on gastrointestinal bacteria. Two such antibodies appeared in primates in recent geological times. Anti-Gal, abundant in humans, apes and Old-World monkeys, appeared 20-30 million years ago (mya) following inactivation of the α1,3GT gene (GGTA1). This gene encodes in other mammals the enzyme α1,3galactosyltransferase (α1,3GT) that synthesizes α-gal epitopes (Galα1-3Galβ1-4GlcNAc-R) which bind anti-Gal. Anti-Neu5Gc, found only in humans, appeared in hominins <6 mya, following elimination of N-glycolylneuraminic-acid (Neu5Gc) because of inactivation of CMAH, the gene encoding hydroxylase that converts N-acetylneuraminic-acid (Neu5Ac) into Neu5Gc. These antibodies, were initially produced in few individuals that acquired random mutations inactivating the corresponding genes and eliminating α-gal epitopes or Neu5Gc, which became nonself antigens. It is suggested that these evolutionary selection events were induced by epidemics of enveloped viruses, lethal to ancestral Old World primates or hominins. Such viruses presented α-gal epitopes or Neu5Gc, synthesized in primates that conserved active GGTA1 or CMAH, respectively, and were lethal to their hosts. The natural anti-Gal or anti-Neu5Gc antibodies, produced in offspring lacking the corresponding carbohydrate antigens, neutralized and destroyed viruses presenting α-gal epitopes or Neu5Gc. These antibodies further induced rapid, effective immune responses against virus antigens, thus preventing infections from reaching lethal stages. These epidemics ultimately resulted in extinction of primate populations synthesizing these carbohydrate antigens and their replacement with offspring populations lacking the antigens and producing protective antibodies against them. Similar events could mediate the elimination of various carbohydrate antigens, thus preventing the complete extinction of other vertebrate species.
Collapse
Affiliation(s)
- Uri Galili
- University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
25
|
Platz J, Bonenfant NR, Uhl FE, Coffey AL, McKnight T, Parsons C, Sokocevic D, Borg ZD, Lam YW, Deng B, Fields JG, DeSarno M, Loi R, Hoffman AM, Bianchi J, Dacken B, Petersen T, Wagner DE, Weiss DJ. Comparative Decellularization and Recellularization of Wild-Type and Alpha 1,3 Galactosyltransferase Knockout Pig Lungs: A Model for Ex Vivo Xenogeneic Lung Bioengineering and Transplantation. Tissue Eng Part C Methods 2016; 22:725-39. [PMID: 27310581 DOI: 10.1089/ten.tec.2016.0109] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND A novel potential approach for lung transplantation could be to utilize xenogeneic decellularized pig lung scaffolds that are recellularized with human lung cells. However, pig tissues express several immunogenic proteins, notably galactosylated cell surface glycoproteins resulting from alpha 1,3 galactosyltransferase (α-gal) activity, that could conceivably prevent effective use. Use of lungs from α-gal knock out (α-gal KO) pigs presents a potential alternative and thus comparative de- and recellularization of wild-type and α-gal KO pig lungs was assessed. METHODS Decellularized lungs were compared by histologic, immunohistochemical, and mass spectrometric techniques. Recellularization was assessed following compartmental inoculation of human lung bronchial epithelial cells, human lung fibroblasts, human bone marrow-derived mesenchymal stromal cells (all via airway inoculation), and human pulmonary vascular endothelial cells (CBF) (vascular inoculation). RESULTS No obvious differences in histologic structure was observed but an approximate 25% difference in retention of residual proteins was determined between decellularized wild-type and α-gal KO pig lungs, including retention of α-galactosylated epitopes in acellular wild-type pig lungs. However, robust initial recellularization and subsequent growth and proliferation was observed for all cell types with no obvious differences between cells seeded into wild-type versus α-gal KO lungs. CONCLUSION These proof of concept studies demonstrate that decellularized wild-type and α-gal KO pig lungs can be comparably decellularized and comparably support initial growth of human lung cells, despite some differences in retained proteins. α-Gal KO pig lungs are a suitable platform for further studies of xenogeneic lung regeneration.
Collapse
Affiliation(s)
- Joseph Platz
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont
| | - Nicholas R Bonenfant
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont
| | - Franziska E Uhl
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont
| | - Amy L Coffey
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont
| | - Tristan McKnight
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont
| | - Charles Parsons
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont
| | - Dino Sokocevic
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont
| | - Zachary D Borg
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont
| | - Ying-Wai Lam
- 2 Department of Biology and VGN Proteomics Facility, University of Vermont College of Arts and Sciences , Burlington, Vermont
| | - Bin Deng
- 2 Department of Biology and VGN Proteomics Facility, University of Vermont College of Arts and Sciences , Burlington, Vermont
| | - Julia G Fields
- 2 Department of Biology and VGN Proteomics Facility, University of Vermont College of Arts and Sciences , Burlington, Vermont
| | - Michael DeSarno
- 3 Biostatistics Unit, University of Vermont College of Medicine , Burlington, Vermont
| | - Roberto Loi
- 4 Department of Biomedical Sciences, University of Cagliari , Cagliari, Italy
| | - Andrew M Hoffman
- 5 Department of Clinical Sciences, Tufts University , Cummings School of Veterinary Medicine, North Grafton, Massachusetts
| | | | | | - Thomas Petersen
- 8 United Therapeutics Corp., Research Triangle Park , Durham, North Carolina
| | - Darcy E Wagner
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont.,9 Comprehensive Pneumonology Center, Helmholtz Center Munich, Ludwig Maximilians University Munich , Munich, Germany
| | - Daniel J Weiss
- 1 Department of Medicine, University of Vermont College of Medicine , Burlington, Vermont
| |
Collapse
|
26
|
Namiri M, Ashtiani MK, Mashinchian O, Hasani-Sadrabadi MM, Mahmoudi M, Aghdami N, Baharvand H. Engineering natural heart valves: possibilities and challenges. J Tissue Eng Regen Med 2016; 11:1675-1683. [PMID: 26799729 DOI: 10.1002/term.2127] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 11/07/2015] [Accepted: 11/30/2015] [Indexed: 12/23/2022]
Abstract
Heart valve replacement is considered to be the most prevalent treatment approach for cardiac valve-related diseases. Among current solutions for heart valve replacement, e.g. mechanical and bioprosthetic valves, the main shortcoming is the lack of growth capability, repair and remodelling of the substitute valve. During the past three decades, tissue engineering-based approaches have shown tremendous potential to overcome these limitations by the development of a biodegradable scaffold, which provides biomechanical and biochemical properties of the native tissue. Among various scaffolds employed for tissue engineering, the decellularized heart valve (DHV) has attracted much attention, due to its native structure as well as comparable haemodynamic characteristics. Although the human DHV has shown optimal properties for valve replacement, the limitation of valve donors in terms of time and size is their main clinical issue. In this regard, xenogenic DHV can be a promising candidate for heart valve replacement. Xenogenic DHVs have similar composition to human valves, which will overcome the need for human DHVs. The main concern regarding xenogeneic DHV replacement is the immunological reaction and calcification following implantation, weak mechanical properties and insufficient recellularization capacity. In this review, we describe the essential steps required to address these impediments through novel engineering approaches. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Mehrnaz Namiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Mohammad Kazemi Ashtiani
- Department of Stem Cells and Developmental Biology, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Omid Mashinchian
- Department of Stem Cells and Developmental Biology, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Mahdi Hasani-Sadrabadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Bioengineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Morteza Mahmoudi
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, CA, USA.,Department of Nanotechnology and Nanotechnology Research Centre, Tehran University of Medical Sciences, Iran.,Cardiovascular Institute, Stanford University School of Medicine, CA, USA
| | - Nasser Aghdami
- Department of Stem Cells and Developmental Biology, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, ACECR, Tehran, Iran
| |
Collapse
|
27
|
Miyagawa S, Matsunari H, Watanabe M, Nakano K, Umeyama K, Sakai R, Takayanagi S, Takeishi T, Fukuda T, Yashima S, Maeda A, Eguchi H, Okuyama H, Nagaya M, Nagashima H. Generation of α1,3-galactosyltransferase and cytidine monophospho-N-acetylneuraminic acid hydroxylase gene double-knockout pigs. J Reprod Dev 2015; 61:449-57. [PMID: 26227017 PMCID: PMC4623151 DOI: 10.1262/jrd.2015-058] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Zinc-finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs) are new tools for
producing gene knockout (KO) animals. The current study reports produced genetically modified pigs, in which
two endogenous genes were knocked out. Porcine fibroblast cell lines were derived from homozygous
α1,3-galactosyltransferase (GalT) KO pigs. These cells were subjected to an additional KO for
the cytidine monophospho-N-acetylneuraminic acid hydroxylase (CMAH) gene. A
pair of ZFN-encoding mRNAs targeting exon 8 of the CMAH gene was used to generate the
heterozygous CMAH KO cells, from which cloned pigs were produced by somatic cell nuclear
transfer (SCNT). One of the cloned pigs obtained was re-cloned after additional KO of the remaining
CMAH allele using the same ZFN-encoding mRNAs to generate
GalT/CMAH-double homozygous KO pigs. On the other hand, the use of
TALEN-encoding mRNAs targeting exon 7 of the CMAH gene resulted in efficient generation of
homozygous CMAH KO cells. These cells were used for SCNT to produce cloned pigs homozygous
for a double GalT/CMAH KO. These results demonstrate that the combination of
TALEN-encoding mRNA, in vitro selection of the nuclear donor cells and SCNT provides a robust
method for generating KO pigs.
Collapse
Affiliation(s)
- Shuji Miyagawa
- Division of Organ Transplantation, Department of Surgery, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Significance of the evolutionary α1,3-galactosyltransferase (GGTA1) gene inactivation in preventing extinction of apes and old world monkeys. J Mol Evol 2014; 80:1-9. [PMID: 25315716 DOI: 10.1007/s00239-014-9652-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 10/06/2014] [Indexed: 10/24/2022]
Abstract
The α1,3-galactosyltransferase (α1,3GT or GGTA1) gene displays unique evolutionary characteristics. This gene appeared early in mammalian evolution and is absent in other vertebrates. The α1,3GT gene is active in marsupials, nonprimate placental mammals, lemurs (prosimians) and New World monkeys, encoding the α1,3GT enzyme that synthesizes a carbohydrate antigen called "α-gal epitope." The α-gal epitope is present in large numbers on cell membrane glycolipids and glycoproteins. The α1,3GT gene was inactivated in ancestral Old World monkeys and apes by frameshift single-base deletions forming premature stop codons. Because of this gene inactivation, humans, apes, and Old World monkeys lack α-gal epitopes and naturally produce an antibody called the "anti-Gal antibody" which binds specifically to α-gal epitopes and which is the most abundant antibody in humans. The evolutionary event that resulted in the inactivation of the α1,3GT gene in ancestral Old World primates could have been mediated by a pathogen endemic to Eurasia-Africa landmass that exerted pressure for selection of primate populations lacking the α-gal epitope. Once the α-gal epitope was eliminated, primates could produce the anti-Gal antibody, possibly as means of defense against pathogens expressing this epitope. This assumption is supported by the fossil record demonstrating an almost complete extinction of apes in the late Miocene and failure of Old World monkeys to radiate into multiple species before that period. A present outcome of this evolutionary event is the anti-Gal-mediated rejection of mammalian xenografts expressing α-gal epitopes in humans, apes, and Old World monkeys.
Collapse
|
29
|
Burlak C, Paris L, Lutz A, Sidner R, Estrada J, Li P, Tector M, Tector A. Reduced binding of human antibodies to cells from GGTA1/CMAH KO pigs. Am J Transplant 2014; 14:1895-900. [PMID: 24909344 PMCID: PMC4366649 DOI: 10.1111/ajt.12744] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 03/07/2014] [Accepted: 03/12/2014] [Indexed: 01/25/2023]
Abstract
Xenotransplantation using genetically modified pig organs could solve the donor organ shortage problem. Two inactivated genes that make humans unique from pigs are GGTA1 and CMAH, the products of which produce the carbohydrate epitopes, aGal and Neu5Gc that attract preformed human antibody. When the GGTA1 and CMAH genes were deleted in pigs, human antibody binding was reduced in preliminary analysis. We analyzed the binding of human IgM and IgG from 121 healthy human serum samples for binding to GGTA1 KO and GGTA1/CMAH KO peripheral blood mononuclear cells (PBMCs). We analyzed a sub population for reactivity toward genetically modified pig PBMCs as compared to chimpanzee and human PBMCs. Deletion of the GGTA1 and CMAH genes in pigs improved the crossmatch results beyond those observed with chimpanzees. Sorting the 121 human samples tested against the GGTA1/CMAH KO pig PBMCs did not reveal a distinguishing feature such as blood group, age or gender. Modification of genes to make pig carbohydrates more similar to humans has improved the crossmatch with human serum significantly.
Collapse
Affiliation(s)
- C. Burlak
- Indiana University School of Medicine, Department of Surgery, Indianapolis, Indiana
| | - L.L. Paris
- Indiana University School of Medicine, Department of Surgery, Indianapolis, Indiana
| | - A.J. Lutz
- Indiana University School of Medicine, Department of Surgery, Indianapolis, Indiana
| | - R.A. Sidner
- Indiana University School of Medicine, Department of Surgery, Indianapolis, Indiana
| | - J. Estrada
- Indiana University School of Medicine, Department of Surgery, Indianapolis, Indiana
| | - P. Li
- Indiana University School of Medicine, Department of Surgery, Indianapolis, Indiana
| | - M. Tector
- Indiana University Health Transplant Institute, Indianapolis, Indiana
| | - A.J. Tector
- Indiana University Health Transplant Institute, Indianapolis, Indiana
| |
Collapse
|
30
|
Amon R, Reuven EM, Leviatan Ben-Arye S, Padler-Karavani V. Glycans in immune recognition and response. Carbohydr Res 2014; 389:115-22. [PMID: 24680512 DOI: 10.1016/j.carres.2014.02.004] [Citation(s) in RCA: 271] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 01/29/2014] [Accepted: 02/02/2014] [Indexed: 11/16/2022]
Abstract
Glycans at the forefront of cells facilitate immune recognition processes. Cancer cells commonly present altered cell surface glycosylation, especially manifested in the expression of sialic acid at the termini of glycolipids and glycoproteins. Although tumor-associated carbohydrate antigens (TACAs) result in expression of altered-self, most such carbohydrates do not elicit strong humoral responses. Various strategies had been devised to elicit increased immunogenicity of such TACA aiming for potent immunotherapeutic antibodies or cancer vaccines. However some carbohydrates are immunogenic in humans and hold potential for novel glycotherapies. N-Glycolylneuraminic acid (Neu5Gc) is a foreign immunogenic sugar in humans originating from the diet (e.g., red meat) and subsequently expressed on the cell surface, especially accumulating on carcinoma. Consequently, the human immune system detects this non-self carbohydrate generating a broad anti-Neu5Gc antibody response. The co-existence of Neu5Gc/anti-Neu5Gc within humans spurs chronic inflammation mediated disease, including cancer. Concurrently, anti-Neu5Gc antibodies hold potential for novel targeted therapy. αGal is another foreign immunogenic carbohydrate antigen in humans and all humans have circulating anti-Gal antibodies. This review aims to describe the immunogenicity of Neu5Gc and its implications for human diseases, highlighting differences and similarities with αGal and its potential for novel targeted theranostics.
Collapse
Affiliation(s)
- Ron Amon
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Eliran Moshe Reuven
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Shani Leviatan Ben-Arye
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Vered Padler-Karavani
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
31
|
Phanse Y, Carrillo-Conde BR, Ramer-Tait AE, Broderick S, Kong CS, Rajan K, Flick R, Mandell RB, Narasimhan B, Wannemuehler MJ. A systems approach to designing next generation vaccines: combining α-galactose modified antigens with nanoparticle platforms. Sci Rep 2014; 4:3775. [PMID: 24441019 PMCID: PMC3895907 DOI: 10.1038/srep03775] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 12/20/2013] [Indexed: 12/21/2022] Open
Abstract
Innovative vaccine platforms are needed to develop effective countermeasures against emerging and re-emerging diseases. These platforms should direct antigen internalization by antigen presenting cells and promote immunogenic responses. This work describes an innovative systems approach combining two novel platforms, αGalactose (αGal)-modification of antigens and amphiphilic polyanhydride nanoparticles as vaccine delivery vehicles, to rationally design vaccine formulations. Regimens comprising soluble αGal-modified antigen and nanoparticle-encapsulated unmodified antigen induced a high titer, high avidity antibody response with broader epitope recognition of antigenic peptides than other regimen. Proliferation of antigen-specific CD4+ T cells was also enhanced compared to a traditional adjuvant. Combining the technology platforms and augmenting immune response studies with peptide arrays and informatics analysis provides a new paradigm for rational, systems-based design of next generation vaccine platforms against emerging and re-emerging pathogens.
Collapse
Affiliation(s)
- Yashdeep Phanse
- 1] Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50011 [2]
| | - Brenda R Carrillo-Conde
- 1] Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011 [2]
| | - Amanda E Ramer-Tait
- 1] Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50011 [2] Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Scott Broderick
- Department of Materials Science and Engineering, Iowa State University, Ames, IA 50011
| | - Chang Sun Kong
- Department of Materials Science and Engineering, Iowa State University, Ames, IA 50011
| | - Krishna Rajan
- Department of Materials Science and Engineering, Iowa State University, Ames, IA 50011
| | - Ramon Flick
- BioProtection Systems Corporation, a subsidiary of NewLink Genetics Corporation, Ames, IA 50010
| | - Robert B Mandell
- 1] BioProtection Systems Corporation, a subsidiary of NewLink Genetics Corporation, Ames, IA 50010 [2]
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011
| | - Michael J Wannemuehler
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50011
| |
Collapse
|
32
|
Schneider MKJ, Seebach JD. Xenotransplantation literature update, September-October 2013. Xenotransplantation 2013; 20:481-6. [PMID: 24289471 DOI: 10.1111/xen.12076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 10/15/2013] [Indexed: 11/28/2022]
Affiliation(s)
- Mårten K J Schneider
- Laboratory of Vascular Immunology, Division of Internal Medicine, University Hospital Zurich, Zurich, Switzerland
| | | |
Collapse
|