1
|
Galili U. Self-Tumor Antigens in Solid Tumors Turned into Vaccines by α-gal Micelle Immunotherapy. Pharmaceutics 2024; 16:1263. [PMID: 39458595 PMCID: PMC11510312 DOI: 10.3390/pharmaceutics16101263] [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: 08/10/2024] [Revised: 09/02/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
A major reason for the failure of the immune system to detect tumor antigens (TAs) is the insufficient uptake, processing, and presentation of TAs by antigen-presenting cells (APCs). The immunogenicity of TAs in the individual patient can be markedly increased by the in situ targeting of tumor cells for robust uptake by APCs, without the need to identify and characterize the TAs. This is feasible by the intra-tumoral injection of α-gal micelles comprised of glycolipids presenting the carbohydrate-antigen "α-gal epitope" (Galα1-3Galβ1-4GlcNAc-R). Humans produce a natural antibody called "anti-Gal" (constituting ~1% of immunoglobulins), which binds to α-gal epitopes. Tumor-injected α-gal micelles spontaneously insert into tumor cell membranes, so that multiple α-gal epitopes are presented on tumor cells. Anti-Gal binding to these epitopes activates the complement system, resulting in the killing of tumor cells, and the recruitment of multiple APCs (dendritic cells and macrophages) into treated tumors by the chemotactic complement cleavage peptides C5a and C3a. In this process of converting the treated tumor into a personalized TA vaccine, the recruited APC phagocytose anti-Gal opsonized tumor cells and cell membranes, process the internalized TAs and transport them to regional lymph-nodes. TA peptides presented on APCs activate TA-specific T cells to proliferate and destroy the metastatic tumor cells presenting the TAs. Studies in anti-Gal-producing mice demonstrated the induction of effective protection against distant metastases of the highly tumorigenic B16 melanoma following injection of natural and synthetic α-gal micelles into primary tumors. This treatment was further found to synergize with checkpoint inhibitor therapy by the anti-PD1 antibody. Phase-1 clinical trials indicated that α-gal micelle immunotherapy is safe and can induce the infiltration of CD4+ and CD8+ T cells into untreated distant metastases. It is suggested that, in addition to converting treated metastases into an autologous TA vaccine, this treatment should be considered as a neoadjuvant therapy, administering α-gal micelles into primary tumors immediately following their detection. Such an immunotherapy will convert tumors into a personalized anti-TA vaccine for the period prior to their resection.
Collapse
Affiliation(s)
- Uri Galili
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
2
|
Wilson JM, Erickson L, Levin M, Ailsworth SM, Commins SP, Platts-Mills TAE. Tick bites, IgE to galactose-alpha-1,3-galactose and urticarial or anaphylactic reactions to mammalian meat: The alpha-gal syndrome. Allergy 2024; 79:1440-1454. [PMID: 38193233 PMCID: PMC11142869 DOI: 10.1111/all.16003] [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: 07/29/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/10/2024]
Abstract
The recent recognition of a syndrome of tick-acquired mammalian meat allergy has transformed the previously held view that mammalian meat is an uncommon allergen. The syndrome, mediated by IgE antibodies against the oligosaccharide galactose-alpha-1,3-galactose (alpha-gal), can also involve reactions to visceral organs, dairy, gelatin and other products, including medications sourced from non-primate mammals. Thus, fittingly, this allergic disorder is now called the alpha-gal syndrome (AGS). The syndrome is strikingly regional, reflecting the important role of tick bites in sensitization, and is more common in demographic groups at risk of tick exposure. Reactions in AGS are delayed, often by 2-6 h after ingestion of mammalian meat. In addition to classic allergic symptomatology such as urticaria and anaphylaxis, AGS is increasingly recognized as a cause of isolated gastrointestinal morbidity and alpha-gal sensitization has also been linked with cardiovascular disease. The unusual link with tick bites may be explained by the fact that allergic cells and mediators are mobilized to the site of tick bites and play a role in resistance against ticks and tick-borne infections. IgE directed to alpha-gal is likely an incidental consequence of what is otherwise an adaptive immune strategy for host defense against endo- and ectoparasites, including ticks.
Collapse
Affiliation(s)
- Jeffrey M. Wilson
- Division of Allergy and Immunology, University of Virginia, Charlottesville, Virginia, USA
| | - Loren Erickson
- Department of Microbiology, Immunology, and Cancer Biology and Beirne Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, USA
| | | | - Samuel M. Ailsworth
- Division of Allergy and Immunology, University of Virginia, Charlottesville, Virginia, USA
| | - Scott P. Commins
- Division of Rheumatology, Allergy and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| | | |
Collapse
|
3
|
Galili U, Li J, Schaer GL. Regeneration in Mice of Injured Skin, Heart, and Spinal Cord by α-Gal Nanoparticles Recapitulates Regeneration in Amphibians. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:730. [PMID: 38668224 PMCID: PMC11055133 DOI: 10.3390/nano14080730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 04/29/2024]
Abstract
The healing of skin wounds, myocardial, and spinal cord injuries in salamander, newt, and axolotl amphibians, and in mouse neonates, results in scar-free regeneration, whereas injuries in adult mice heal by fibrosis and scar formation. Although both types of healing are mediated by macrophages, regeneration in these amphibians and in mouse neonates also involves innate activation of the complement system. These differences suggest that localized complement activation in adult mouse injuries might induce regeneration instead of the default fibrosis and scar formation. Localized complement activation is feasible by antigen/antibody interaction between biodegradable nanoparticles presenting α-gal epitopes (α-gal nanoparticles) and the natural anti-Gal antibody which is abundant in humans. Administration of α-gal nanoparticles into injuries of anti-Gal-producing adult mice results in localized complement activation which induces rapid and extensive macrophage recruitment. These macrophages bind anti-Gal-coated α-gal nanoparticles and polarize into M2 pro-regenerative macrophages that orchestrate accelerated scar-free regeneration of skin wounds and regeneration of myocardium injured by myocardial infarction (MI). Furthermore, injection of α-gal nanoparticles into spinal cord injuries of anti-Gal-producing adult mice induces recruitment of M2 macrophages, that mediate extensive angiogenesis and axonal sprouting, which reconnects between proximal and distal severed axons. Thus, α-gal nanoparticle treatment in adult mice mimics physiologic regeneration in amphibians. These studies further suggest that α-gal nanoparticles may be of significance in the treatment of human injuries.
Collapse
Affiliation(s)
- Uri Galili
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA; (J.L.); (G.L.S.)
| | | | | |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Montgomery RA, Tang WHW. Cardiac Xenotransplantation: a New Frontier for Advanced Heart Failure. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2023; 25:65-78. [PMID: 38957658 PMCID: PMC11218470 DOI: 10.1007/s11936-023-00977-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/13/2023] [Indexed: 02/11/2023]
Abstract
Purpose of review Cardiac transplantation is a critical treatment for patients with advanced heart failure, offering the ability to markedly improve quality and quantity of life. Unfortunately, this treatment is limited by donor organ availability, despite efforts to increase organ supply and improve donor organ allocation and usage. The transplantation of non-human donor organs (xenotransplantation) offers to readily address many limitations of the current transplantation system; however, scattered attempts to establish this practice have been met with frustration. In this review, we discuss the limitations of the historical attempts and outline recent progress in the field of cardiac xenotransplantation. Recent findings The advent of CRISPR-Cas9 genome editing techniques and emerging commercial and regulatory alignment has led to a flurry of new attempts to establish xenotransplantation as a viable treatment for those with end-stage heart failure. The first xenotransplantation of a genetically modified pig heart to a human recipient on January 7, 2022, highlighted the progress the science of xenotransplantation has made, as well as the need to outline next steps to further establish the practice. Summary The development of a genetically modified porcine model has renewed hope that xenotransplantation might succeed where prior attempts failed, though many barriers remain.
Collapse
Affiliation(s)
- Robert A. Montgomery
- Harrington Heart and Vascular Institute, University Hospitals, Cleveland, OH, USA
| | - W. H. Wilson Tang
- Kaufman Center for Heart Failure Treatment and Recovery, Heart Vascular and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, Desk J3–4, Cleveland, OH 44195, USA
| |
Collapse
|
7
|
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
|
8
|
Yuan B, Tang YF, Xu Z, Wang JC, Zhou SY, Chen XS. Lyophilized bovine acellular tendon linear fiber material for the reconstruction of attachment structure of paraspinous muscles: an animal in vivo study. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2022; 33:79. [PMID: 36462052 PMCID: PMC9719447 DOI: 10.1007/s10856-022-06701-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 11/10/2022] [Indexed: 06/17/2023]
Abstract
Low back pain is common after lumbar spine surgery and the injury from extensive detachment of paraspinal muscles during the surgery may play a vital role. Previously, we prepared a bovine acellular tendon fiber (ATF) material through lyophilization and proved that it could retain its original fibrillar structure and mechanical properties. The objective of this study is to evaluate the effectiveness of this new fiber material used for attachment structure reconstruction of paraspinal muscle. Defect of spinous process, interspinous and supraspinous ligament was established on lumbar spine in rabbit and rat and ATF linear material was implanted to reconstruct the attachment structure. Ultrasound showed the cross-sectional area of the paraspinal muscle in ATF group was larger than that of control group in rats. MRI showed the irregular shape and high signal changes in control group, but regular shape and uniform signal in the ATF group in rabbit. For Electromyogram, the frequency of evoked potential in control group was lower than ATF group and normal rats. HE and Masson staining showed good tissue healing, and immunohistochemical results showed the immune rejection of ATF is significantly lower than that of suture. Reconstruction of the attachment structure of paraspinous muscles with ATF linear material could maintain the morphology, volume and function of paraspinal muscle. ATF material has the potential to be used to manufacture personalized ligaments and other tissue engineering scaffolds. Graphical abstract.
Collapse
Affiliation(s)
- Bo Yuan
- Spine Center, Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Yi-Fan Tang
- Spine Center, Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Zheng Xu
- Spine Center, Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Jun-Cheng Wang
- Spine Center, Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Sheng-Yuan Zhou
- Spine Center, Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China.
| | - Xiong-Sheng Chen
- Spine Center, Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China.
| |
Collapse
|
9
|
de Lederkremer RM, Giorgi ME, Marino C. The α-Galactosyl Carbohydrate Epitope in Pathogenic Protozoa. ACS Infect Dis 2022; 8:2207-2222. [PMID: 36083842 DOI: 10.1021/acsinfecdis.2c00370] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The α-gal epitope, which refers to the carbohydrate α-d-Galp-(1 → 3)-β-d-Galp-(1 → 4)-d-GlcNAc-R, was first described in the glycoconjugates of mammals other than humans. Evolution caused a mutation that resulted in the inactivation of the α-1,3-galactosyltransferase gene. For that reason, humans produce antibodies against α-d-Galp containing glycoproteins and glycolipids of other species. We summarize here the glycoconjugates with α-d-Galp structures in Trypanosoma, Leishmania, and Plasmodium pathogenic protozoa. These were identified in infective stages of Trypanosoma cruzi and in Plasmodium sporozoites. In Leishmania, α-d-Galp is linked differently in the glycans of glycoinositolphospholipids (GIPLs). Chemically synthesized neoglycoconjugates have been proposed as diagnostic tools and as antigens for vaccines. Several syntheses reported for the α-gal trisaccharide, also called the Galili epitope, and the glycans of GIPLs found in Leishmania, the preparation of neoglycoconjugates, and the studies in which they were involved are also included in this Review.
Collapse
Affiliation(s)
- Rosa M de Lederkremer
- CIHIDECAR, Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Pabellón II, Ciudad Universitaria, 1428Buenos Aires, Argentina
| | - María Eugenia Giorgi
- CIHIDECAR, Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Pabellón II, Ciudad Universitaria, 1428Buenos Aires, Argentina
| | - Carla Marino
- CIHIDECAR, Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Pabellón II, Ciudad Universitaria, 1428Buenos Aires, Argentina
| |
Collapse
|
10
|
GTKO rabbit: A novel animal model for preclinical assessment of decellularized xenogeneic grafts via in situ implantation. Mater Today Bio 2022; 18:100505. [DOI: 10.1016/j.mtbio.2022.100505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 11/06/2022] [Accepted: 11/24/2022] [Indexed: 11/26/2022]
|
11
|
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
|
12
|
Lopez KJ, Cross-Najafi AA, Farag K, Obando B, Thadasina D, Isidan A, Park Y, Zhang W, Ekser B, Li P. Strategies to induce natural killer cell tolerance in xenotransplantation. Front Immunol 2022; 13:941880. [PMID: 36072599 PMCID: PMC9441937 DOI: 10.3389/fimmu.2022.941880] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/28/2022] [Indexed: 11/23/2022] Open
Abstract
Eliminating major xenoantigens in pig cells has drastically reduced human antibody-mediated hyperacute xenograft rejection (HXR). Despite these advancements, acute xenograft rejection (AXR) remains one of the major obstacles to clinical xenotransplantation, mediated by innate immune cells, including macrophages, neutrophils, and natural killer (NK) cells. NK cells play an ‘effector’ role by releasing cytotoxicity granules against xenogeneic cells and an ‘affecter’ role on other immune cells through cytokine secretion. We highlight the key receptor-ligand interactions that determine the NK cell response to target cells, focusing on the regulation of NK cell activating receptor (NKG2D, DNAM1) and inhibitory receptor (KIR2DL1-4, NKG2A, and LIR-1) signaling pathways. Inhibition of NK cell activity may protect xenografts from cytotoxicity. Recent successful approaches to reducing NK cell-mediated HXR and AXR are reviewed, including genetic modifications of porcine xenografts aimed at improving pig-to-human compatibility. Future directions to promote xenograft acceptance are discussed, including NK cell tolerance in pregnancy and NK cell evasion in viral infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ping Li
- *Correspondence: Ping Li, ; Burcin Ekser,
| |
Collapse
|
13
|
Campos-Sánchez JC, Guardiola FA, Esteban MÁ. In vitro effects of λ-carrageenin in the head-kidney leucocytes of gilthead seabream (Sparus aurata). FISH & SHELLFISH IMMUNOLOGY 2022; 127:813-821. [PMID: 35842113 DOI: 10.1016/j.fsi.2022.07.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 06/30/2022] [Accepted: 07/11/2022] [Indexed: 06/15/2023]
Abstract
The λ-carrageenin is a sulphated mucopolysaccharide that has been used for decades to induce experimental inflammation in mammals. However, it has been little considered in fish. We studied the in vitro effects of λ-carrageenin on gilthead seabream (Sparus aurata L.) head-kidney leucocytes (HKLs). For this purpose, HKLs were incubated with serial concentrations (from 0 to 1,000 μg mL-1) of λ-carrageenin for 3, 6, 12 and 24 h to assess its influence on cell viability and morphology, cell activity and modulation of several selected inflammation-related genes. The viability results demonstrated that λ-carrageenin has no negative effects on HKLs. The respiratory burst activity and phagocytic ability of HKLs after being incubated with λ-carrageenin (100 and 1,000 μg mL-1) for 24 h were increased, whereas the phagocytic capacity was inhibited by the higher dose at the same experimental time compared with control samples. However, the peroxidase activity of HKLs was not changed by incubation with λ-carrageenin. According to transmission electron microscopy results, incubation of HKLs with the higher dose of λ-carrageenin appeared to activate the cells being evident different morphological changes without sign of cell death. Furthermore, up-regulation of three proinflammatory cytokines (il1b, tnfa, and il6) and down-regulation of anti-inflammatory genes (tgfb) were denoted in HKLs incubated with carrageenin. The present results provide a detailed approach to the effects of λ-carrageenin on fish leucocytes, which could have some impact on how we understand the response of these cells when inducing an inflammatory process in fish.
Collapse
Affiliation(s)
- Jose Carlos Campos-Sánchez
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain
| | - Francisco A Guardiola
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain
| | - María Ángeles Esteban
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain.
| |
Collapse
|
14
|
Langley DB, Schofield P, Nevoltris D, Jackson J, Jackson KJL, Peters TJ, Burk M, Matthews JM, Basten A, Goodnow CC, van Nunen S, Reed JH, Christ D. Genetic and structural basis of the human anti-α-galactosyl antibody response. Proc Natl Acad Sci U S A 2022; 119:e2123212119. [PMID: 35867757 PMCID: PMC9282431 DOI: 10.1073/pnas.2123212119] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/10/2022] [Indexed: 01/11/2023] Open
Abstract
Humans lack the capacity to produce the Galα1-3Galβ1-4GlcNAc (α-gal) glycan, and produce anti-α-gal antibodies upon exposure to the carbohydrate on a diverse set of immunogens, including commensal gut bacteria, malaria parasites, cetuximab, and tick proteins. Here we use X-ray crystallographic analysis of antibodies from α-gal knockout mice and humans in complex with the glycan to reveal a common binding motif, centered on a germline-encoded tryptophan residue at Kabat position 33 (W33) of the complementarity-determining region of the variable heavy chain (CDRH1). Immunoglobulin sequencing of anti-α-gal B cells in healthy humans and tick-induced mammalian meat anaphylaxis patients revealed preferential use of heavy chain germline IGHV3-7, encoding W33, among an otherwise highly polyclonal antibody response. Antigen binding was critically dependent on the presence of the germline-encoded W33 residue for all of the analyzed antibodies; moreover, introduction of the W33 motif into naive IGHV3-23 antibody phage libraries enabled the rapid selection of α-gal binders. Our results outline structural and genetic factors that shape the human anti-α-galactosyl antibody response, and provide a framework for future therapeutics development.
Collapse
Affiliation(s)
- David B. Langley
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Peter Schofield
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Damien Nevoltris
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Jennifer Jackson
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | | | - Tim J. Peters
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Melanie Burk
- Tick-induced Allergies Research and Awareness Centre, Sydney, NSW 2065, Australia
| | - Jacqueline M. Matthews
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Antony Basten
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Christopher C. Goodnow
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
- Cellular Genomics Futures Institute, University of New South Wales, Sydney, NSW 2052, Australia
| | - Sheryl van Nunen
- Tick-induced Allergies Research and Awareness Centre, Sydney, NSW 2065, Australia
- Northern Clinical School, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2065, Australia
| | - Joanne H. Reed
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| | - Daniel Christ
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2010, Australia
| |
Collapse
|
15
|
Zhou SY, Yuan B, Huang WM, Chen XS, Jia LS. Aponeurosis discission, a low-detergent method for tissue-engineered acellular ligament scaffolds. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2022; 33:40. [PMID: 35507049 PMCID: PMC9068632 DOI: 10.1007/s10856-022-06661-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 04/05/2022] [Indexed: 06/14/2023]
Abstract
Detergent treatment is the most commonly used method for the decellularization of ligaments and tendon grafts. However, it is well recognized that detergent treatment can also adversely affect the extracellular matrix. This study found that discission into the aponeurosis layer of the patellar tendon (PT) before decellularization is conducive to extracting cells from the PT using a low quantity of detergent in a short time period. The acellular aponeurosis discission ligament (AADL) retains its native collagen fibril structure and mechanical properties. Moreover, the PT retained cell and tissue compatibility in vitro and in vivo. After implantation into a defective allogeneic PT, we found that the AADL healed well in the host, and its collagen structure exhibited gradual improvement 12 months after implantation with satisfactory reconstruction. IMPACT: The aponeurosis of tendons/ligaments is the main barrier to achieving complete decellularization, and it thus prevents complete recellularization for applications in tissue engineering. Aponeurosis can obstruct the removal of cell components. We found that excising the aponeurosis before decellularization allows for the removal of cellular components with a reduced amount of detergent, thus improving the biological properties of the acellular ligament. To the best of our knowledge, no similar studies have been performed. Graphical abstract.
Collapse
Affiliation(s)
- Sheng-Yuan Zhou
- Spine Center, Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Bo Yuan
- Spine Center, Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Wen-Mao Huang
- Spine Center, Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Xiong-Sheng Chen
- Spine Center, Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China.
| | - Lian-Shun Jia
- Spine Center, Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| |
Collapse
|
16
|
Chakrapani N, Fischer J, Swiontek K, Codreanu-Morel F, Hannachi F, Morisset M, Mugemana C, Bulaev D, Blank S, Bindslev-Jensen C, Biedermann T, Ollert M, Hilger C. α-Gal present on both glycolipids and glycoproteins contributes to immune response in meat-allergic patients. J Allergy Clin Immunol 2022; 150:396-405.e11. [PMID: 35459547 DOI: 10.1016/j.jaci.2022.02.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 02/10/2022] [Accepted: 02/21/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND The α-Gal syndrome is associated with the presence of IgE directed to the carbohydrate galactose-α-1,3-galactose (α-Gal) and is characterized by a delayed allergic reaction occurring 2 to 6 hours after ingestion of mammalian meat. On the basis of their slow digestion and processing kinetics, α-Gal-carrying glycolipids have been proposed as the main trigger of the delayed reaction. OBJECTIVE We analyzed and compared the in vitro allergenicity of α-Gal-carrying glycoproteins and glycolipids from natural food sources. METHODS Proteins and lipids were extracted from pork kidney (PK), beef, and chicken. Glycolipids were purified from rabbit erythrocytes. The presence of α-Gal and IgE binding of α-Gal-allergic patient sera (n = 39) was assessed by thin-layer chromatography as well as by direct and inhibition enzyme-linked immunosorbent assay. The in vitro allergenicity of glycoproteins and glycolipids from different meat extracts was determined by basophil activation test. Glycoprotein stability was evaluated by simulated gastric and intestinal digestion assays. RESULTS α-Gal was detected on glycolipids of PK and beef. Patient IgE antibodies recognized α-Gal bound to glycoproteins and glycolipids, although binding to glycoproteins was more potent. Rabbit glycolipids were able to strongly activate patient basophils, whereas lipid extracts from PK and beef were also found to trigger basophil activation, but at a lower capacity compared to the respective protein extracts. Simulated gastric digestion assays of PK showed a high stability of α-Gal-carrying proteins in PK. CONCLUSION Both α-Gal-carrying glycoproteins and glycolipids are able to strongly activate patient basophils. In PK and beef, α-Gal epitopes seem to be less abundant on glycolipids than on glycoproteins, suggesting a major role of glycoproteins in delayed anaphylaxis upon consumption of these food sources.
Collapse
Affiliation(s)
- Neera Chakrapani
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jörg Fischer
- Department of Dermatology, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Kyra Swiontek
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | | | - Farah Hannachi
- Immunology-Allergology Unit, Centre Hospitalier Luxembourg, Differdange, Luxembourg
| | - Martine Morisset
- Immunology-Allergology Unit, Centre Hospitalier Luxembourg, Differdange, Luxembourg
| | - Clément Mugemana
- Department of Materials Research and Technology, Luxembourg Institute of Science and Technology (LIST), Esch-sur-Alzette, Luxembourg
| | - Dmitry Bulaev
- Competence Center for Methodology and Statistics, LIH, Esch-sur-Alzette, Luxembourg
| | - Simon Blank
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environment Health, Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Carsten Bindslev-Jensen
- Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, Odense, Denmark
| | - Tilo Biedermann
- Department of Dermatology and Allergy Biederstein, Technical University of Munich, Munich, Germany
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg; Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, Odense, Denmark
| | - Christiane Hilger
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
17
|
Olivera-Ardid S, Bello-Gil D, Tuzikov A, Araujo RN, Ferrero-Alves Y, García Figueroa BE, Labrador-Horrillo M, García-Pérez AL, Bovin N, Mañez R. Poly-L-Lysine-Based αGal-Glycoconjugates for Treating Anti-αGal IgE-Mediated Diseases. Front Immunol 2022; 13:873019. [PMID: 35432370 PMCID: PMC9009260 DOI: 10.3389/fimmu.2022.873019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/02/2022] [Indexed: 11/25/2022] Open
Abstract
Anti-αGal IgE antibodies mediate a spreading allergic condition known as αGal-syndrome (AGS). People exposed to hard tick bites are sensitized to αGal, producing elevated levels of anti-αGal IgE, which are responsible for AGS. This work presents an immunotherapy based on polymeric αGal-glycoconjugates for potentially treating allergic disorders by selectively inhibiting anti-αGal IgE antibodies. We synthesized a set of αGal-glycoconjugates, based on poly-L-lysine of different degrees of polymerization (DP1000, DP600, and DP100), to specifically inhibit in vitro the anti-αGal IgE antibodies in the serum of αGal-sensitized patients (n=13). Moreover, an animal model for αGal sensitization in GalT-KO mice was developed by intradermal administration of hard tick' salivary gland extract, mimicking the sensitization mechanism postulated in humans. The in vitro exposure to all polymeric glycoconjugates (5-10-20-50-100 µg/mL) mainly inhibited anti-αGal IgE and IgM isotypes, with a lower inhibition effect on the IgA and IgG, respectively. We demonstrated a differential anti-αGal isotype inhibition as a function of the length of the poly-L-lysine and the number of αGal residues exposed in the glycoconjugates. These results defined a minimum of 27 αGal residues to inhibit most of the induced anti-αGal IgE in vitro. Furthermore, the αGal-glycoconjugate DP1000-RA0118 (10 mg/kg sc.) showed a high capacity to remove the anti-αGal IgE antibodies (≥75% on average) induced in GalT-KO mice, together with similar inhibition for circulating anti-αGal IgG and IgM. Our study suggests the potential clinical use of poly-L-lysine-based αGal-glycoconjugates for treating allergic disorders mediated by anti-αGal IgE antibodies.
Collapse
Affiliation(s)
- Sara Olivera-Ardid
- RemAb Therapeutics, Mòdul de Recerca B, UAB Bellaterra, Barcelona, Spain
| | - Daniel Bello-Gil
- RemAb Therapeutics, Mòdul de Recerca B, UAB Bellaterra, Barcelona, Spain
| | - Alexander Tuzikov
- Department of Chemical Biology of Glycans and Lipids, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences (RAS), Moscow, Russia
| | - Ricardo N. Araujo
- Laboratório de Artrópodes Hematófagos, Departamento de Parasitologia, ICB/UFMG, Belo Horizonte, Brazil
| | - Yara Ferrero-Alves
- RemAb Therapeutics, Mòdul de Recerca B, UAB Bellaterra, Barcelona, Spain
| | - Blanca Esther García Figueroa
- MEGA: Asthma Inception and Progression Mechanisms, Complejo Hospitalario de Navarra (CHN), Pamplona, Spain
- Instituto de investigación sanitaria de Navarra (IdiSNA), Pamplona, Spain
- ARADyAL Research Network, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Moisés Labrador-Horrillo
- ARADyAL Research Network, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Allergy Section, Internal Medicine Department, Hospital Universitari Vall d’Hebron (HUVH), Barcelona, Spain
- Immunomediated Diseases and Innovative Therapies, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Ana L. García-Pérez
- Departamento de Sanidad Animal, Instituto Vasco de Investigación de Desarrollo Agrario (NEIKER), Derio, Spain
| | - Nicolai Bovin
- Department of Chemical Biology of Glycans and Lipids, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences (RAS), Moscow, Russia
| | - Rafael Mañez
- RemAb Therapeutics, Mòdul de Recerca B, UAB Bellaterra, Barcelona, Spain
- Hospital Universitari de Bellvitge, Servicio de Medicina Intensiva, Hospitalet de Llobregat, Barcelona, Spain
- Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Grupo Inmunidad Innata y Patología del Paciente Crítico, Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
18
|
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
|
19
|
Zhou S, Yuan B, Huang W, Tang Y, Chen X. Preparation and biological characteristics of a bovine acellular tendon fiber material. J Biomed Mater Res A 2021; 109:1931-1941. [PMID: 33811434 DOI: 10.1002/jbm.a.37185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 03/14/2021] [Accepted: 03/24/2021] [Indexed: 11/07/2022]
Abstract
Acellular tendon matrix is an ideal substitute for constructing tissue engineering ligaments, but using detergents causes damage to collagen and fibrin during the process of decellularization. In this study, fresh tendons were lyophilized and separated into fresh tendon fiber (FTF) bundles, and then the cellular components in FTF were removed to prepare acellular tendon fiber (ATF) without adding chemical detergent. H&E staining and DAPI fluorescence microscopy showed no nucleus and DNA residue. Compared with FTFs, the DNA content of ATFs was significantly lower without the collagen content change before and after decellularization. The microstructure of collagen fibrils in ATFs was intact under scanning electron microscopy (SEM), and the maximum tensile load and elastic modulus between FTFs and ATFs were not statistically different. The ATF bundles were cultured with SD rat tenocytes for 72 hr and cells attachment to fiber surfaces were observed under SEM. ATF bundles were then implanted into paraspinal muscles, and histological analysis showed fibroblast-like cells within the ATFs and was similar to the control group (fresh tendon autograft) in morphology. H&E staining showed that the number of lymphocytes and plasma cells in ATF was less than that in fresh tendon autograft. ATF bundles were twisted into linear fiber materials by hand, of which the maximum breaking strength was similar to silk with same diameter. These findings demonstrated that ATFs retain their original fibril structure and mechanical properties after decellularization by trypsin and pancreatic deoxyribonuclease without detergent. Lyophilized ATFs linear fiber material provides the possibility of preparing personalized ligament and other tissue engineering scaffolds.
Collapse
Affiliation(s)
- Shengyuan Zhou
- Spine Center, Department of Orthopedic Surgery, Chang Zheng Hospital, Naval Medical Univeristy (Second Military Medical University), Shanghai, China
| | - Bo Yuan
- Spine Center, Department of Orthopedic Surgery, Chang Zheng Hospital, Naval Medical Univeristy (Second Military Medical University), Shanghai, China
| | - Wenmao Huang
- Spine Center, Department of Orthopedic Surgery, Chang Zheng Hospital, Naval Medical Univeristy (Second Military Medical University), Shanghai, China
| | - Yifan Tang
- Spine Center, Department of Orthopedic Surgery, Chang Zheng Hospital, Naval Medical Univeristy (Second Military Medical University), Shanghai, China
| | - Xiongsheng Chen
- Spine Center, Department of Orthopedic Surgery, Chang Zheng Hospital, Naval Medical Univeristy (Second Military Medical University), Shanghai, China
| |
Collapse
|
20
|
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
|
21
|
Galili U. COVID-19 variants as moving targets and how to stop them by glycoengineered whole-virus vaccines. Virulence 2021; 12:1717-1720. [PMID: 34304693 PMCID: PMC8346202 DOI: 10.1080/21505594.2021.1939924] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Uri Galili
- Department of Medicine, Rush Medical College, Chicago, IL, USA
| |
Collapse
|
22
|
Galili U. Increasing Efficacy of Enveloped Whole-Virus Vaccines by In situ Immune-Complexing with the Natural Anti-Gal Antibody. MEDICAL RESEARCH ARCHIVES 2021; 9:2481. [PMID: 34853815 PMCID: PMC8631339 DOI: 10.18103/mra.v9i7.2481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The appearance of variants of mutated virus in course of the Covid-19 pandemic raises concerns regarding the risk of possible formation of variants that can evade the protective immune response elicited by the single antigen S-protein gene-based vaccines. This risk may be avoided by inclusion of several antigens in vaccines, so that a variant that evades the immune response to the S-protein of SARS-CoV-2 virus will be destroyed by the protective immune response against other viral antigens. A simple way for preparing multi-antigenic enveloped-virus vaccines is using the inactivated whole-virus as vaccine. However, immunogenicity of such vaccines may be suboptimal because of poor uptake of the vaccine by antigen-presenting-cells (APC) due to electrostatic repulsion by the negative charges of sialic-acid on both the glycan-shield of the vaccinating virus and on the carbohydrate-chains (glycans) of APC. In addition, glycan-shield can mask many antigenic peptides. These effects of the glycan-shield can be reduced and immunogenicity of the vaccinating virus markedly increased by glycoengineering viral glycans for replacing sialic-acid units on glycans with α-gal epitopes (Galα1-3Galβ1-4GlcNAc-R). Vaccination of humans with inactivated whole-virus presenting α-gal epitopes (virusα-gal) results in formation of immune-complexes with the abundant natural anti-Gal antibody that binds to viral α-gal epitopes at the vaccination site. These immune-complexes are targeted to APC for rigorous uptake due to binding of the Fc portion of immunecomplexed anti-Gal to Fcγ receptors on APC. The APC further transport the large amounts of internalized vaccinating virus to regional lymph nodes, process and present the virus antigenic peptides for the activation of many clones of virus specific helper and cytotoxic T-cells. This elicits a protective cellular and humoral immune response against multiple viral antigens and an effective immunological memory. The immune response to virusα-gal vaccine was studied in mice producing anti-Gal and immunized with inactivated influenza-virusα-gal. These mice demonstrated 100-fold increase in titer of the antibodies produced, a marked increase in T-cell response, and a near complete protection against challenge with a lethal dose of live influenza-virus, in comparison to a similar vaccine lacking α-gal epitopes. This glycoengineering can be achieved in vitro by enzymatic reaction with neuraminidase removing sialic-acid and with recombinant α1,3galactosyltransferase (α1,3GT) synthesizing α-gal epitopes, by engineering host-cells to contain several copies of the α1,3GT gene (GGTA1), or by transduction of this gene in a replication-defective adenovirus vector into host-cells. Theoretically, these methods for increased immunogenicity may be applicable to all enveloped viruses with N-glycans on their envelope.
Collapse
Affiliation(s)
- Uri Galili
- Department of Medicine, Rush Medical College, Chicago, IL, USA
| |
Collapse
|
23
|
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
|
24
|
Weber P, Fischer R, Nasseri SA, Stütz AE, Thonhofer M, Withers SG, Wolfsgruber A, Wrodnigg TM. New α-galactosidase-inhibiting aminohydroxycyclopentanes. RSC Adv 2021; 11:15943-15951. [PMID: 35481199 PMCID: PMC9029992 DOI: 10.1039/d1ra02507d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/22/2021] [Indexed: 01/09/2023] Open
Abstract
A set of cyclopentanoid α-galactosidase ligands was prepared from a partially protected ω-eno-aldose via a reliable (2 + 3)-cycloaddition protocol with slightly modified conditions. The obtained N-benzylisoxazolidine ring was selectively opened and the configuration of the hydroxymethylgroup was inverted. Consecutive deprotection provided an aminocyclopentane, which was N-alkylated to furnish a set of potential α-galactosidase inhibitors. Their glycosidase inhibitory activities were screened with a panel of standard glycosidases of biological significance. A concise and robust synthesis of new cyclopentanoid competitive inhibitors of α-galactosidases related to Fabry's disease and other α-galactosidase related disorders.![]()
Collapse
Affiliation(s)
- Patrick Weber
- Glycogroup
- Institute of Chemistry and Technology of Biobased Systems
- Graz University of Technology
- A-8010 Graz
- Austria
| | - Roland Fischer
- Institute of Inorganic Chemistry
- Graz University of Technology
- A-8010 Graz
- Austria
| | - Seyed A. Nasseri
- Chemistry Department
- University of British Columbia
- Vancouver
- V6T 1Z1 Canada
| | - Arnold E. Stütz
- Glycogroup
- Institute of Chemistry and Technology of Biobased Systems
- Graz University of Technology
- A-8010 Graz
- Austria
| | - Martin Thonhofer
- Glycogroup
- Institute of Chemistry and Technology of Biobased Systems
- Graz University of Technology
- A-8010 Graz
- Austria
| | - Stephen G. Withers
- Chemistry Department
- University of British Columbia
- Vancouver
- V6T 1Z1 Canada
| | - Andreas Wolfsgruber
- Glycogroup
- Institute of Chemistry and Technology of Biobased Systems
- Graz University of Technology
- A-8010 Graz
- Austria
| | - Tanja M. Wrodnigg
- Glycogroup
- Institute of Chemistry and Technology of Biobased Systems
- Graz University of Technology
- A-8010 Graz
- Austria
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Kappler K, Hennet T. Emergence and significance of carbohydrate-specific antibodies. Genes Immun 2020; 21:224-239. [PMID: 32753697 PMCID: PMC7449879 DOI: 10.1038/s41435-020-0105-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/14/2020] [Accepted: 07/22/2020] [Indexed: 12/14/2022]
Abstract
Carbohydrate-specific antibodies are widespread among all classes of immunoglobulins. Despite their broad occurrence, little is known about their formation and biological significance. Carbohydrate-specific antibodies are often classified as natural antibodies under the assumption that they arise without prior exposure to exogenous antigens. On the other hand, various carbohydrate-specific antibodies, including antibodies to ABO blood group antigens, emerge after the contact of immune cells with the intestinal microbiota, which expresses a vast diversity of carbohydrate antigens. Here we explore the development of carbohydrate-specific antibodies in humans, addressing the definition of natural antibodies and the production of carbohydrate-specific antibodies upon antigen stimulation. We focus on the significance of the intestinal microbiota in shaping carbohydrate-specific antibodies not just in the gut, but also in the blood circulation. The structural similarity between bacterial carbohydrate antigens and surface glycoconjugates of protists, fungi and animals leads to the production of carbohydrate-specific antibodies protective against a broad range of pathogens. Mimicry between bacterial and human glycoconjugates, however, can also lead to the generation of carbohydrate-specific antibodies that cross-react with human antigens, thereby contributing to the development of autoimmune disorders.
Collapse
Affiliation(s)
| | - Thierry Hennet
- Institute of Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
28
|
GGTA1/iGb3S Double Knockout Mice: Immunological Properties and Immunogenicity Response to Xenogeneic Bone Matrix. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9680474. [PMID: 32596401 PMCID: PMC7292995 DOI: 10.1155/2020/9680474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 12/05/2022]
Abstract
Background Animal tissues and tissue-derived biomaterials are widely used in the field of xenotransplantation and regenerative medicine. A potential immunogenic risk that affects the safety and effectiveness of xenografts is the presence of remnant α-Gal antigen (synthesized by GGTA1 or/and iGb3S). GGTA1 knockout mice have been developed as a suitable model for the analysis of anti-Gal antibody-mediated immunogenicity. However, we are yet to establish whether GGTA1/iGb3S double knockout (G/i DKO) mice are sensitive to Gal antigen-positive xenoimplants. Methods α-Gal antigen expression in the main organs of G/i DKO mice or bovine bone substitutes was detected via a standardized ELISA inhibition assay. Serum anti-α-Gal antibody titers of G/i DKO mice after immunization with rabbit red blood cells (RRBC) and implantation of raw lyophilized bone substitutes (Gal antigen content was 8.14 ± 3.17 × 1012/mg) or Guanhao Biotech bone substitutes (50% decrease in Gal antigen relative to the raw material) were assessed. The evaluation of total serum antibody, inflammatory cytokine, and splenic lymphocyte subtype populations and the histological analysis of implants and thymus were performed to systematically assess the immune response caused by bovine bone substitutes and bone substitute grafts in G/i DKO mice. Results α-Gal epitope expression was reduced by 100% in the main organs of G/i DKO mice, compared with their wild-type counterparts. Following immunization with RRBC, serum anti-Gal antibody titers of G/i DKO mice increased from 80- to 180-fold. After subcutaneous implantation of raw lyophilized bone substitutes and Guanhao Biotech bone substitutes into G/i DKO mice, specific anti-α-Gal IgG, anti-α-Gal IgM, and related inflammatory factors (IFN-γ and IL-6) were significantly increased in the raw lyophilized bone substitute group but showed limited changes in the Guanhao Biotech bone substitute group, compared with the control. Conclusion G/i DKO mice are sensitive to Gal antigen-positive xenogeneic grafts and can be effectively utilized for evaluating the α-Gal-mediated immunogenic risk of xenogeneic grafts.
Collapse
|
29
|
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
|
30
|
Chen P, Liu R, Huang M, Zhu J, Wei D, Castellino FJ, Dang G, Xie F, Li G, Cui Z, Liu S, Zhang Y. A unique combination of glycoside hydrolases in Streptococcus suis specifically and sequentially acts on host-derived αGal-epitope glycans. J Biol Chem 2020; 295:10638-10652. [PMID: 32518157 DOI: 10.1074/jbc.ra119.011977] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 06/06/2020] [Indexed: 01/02/2023] Open
Abstract
Infections by many bacterial pathogens rely on their ability to degrade host glycans by producing glycoside hydrolases (GHs). Here, we discovered a conserved multifunctional GH, SsGalNagA, containing a unique combination of two family 32 carbohydrate-binding modules (CBM), a GH16 domain and a GH20 domain, in the zoonotic pathogen Streptococcus suis 05ZYH33. Enzymatic assays revealed that the SsCBM-GH16 domain displays endo-(β1,4)-galactosidase activity specifically toward the host-derived αGal epitope Gal(α1,3)Gal(β1,4)Glc(NAc)-R, whereas the SsGH20 domain has a wide spectrum of exo-β-N-acetylhexosaminidase activities, including exo-(β1,3)-N-acetylglucosaminidase activity, and employs this activity to act in tandem with SsCBM-GH16 on the αGal-epitope glycan. Further, we found that the CBM32 domain adjacent to the SsGH16 domain is indispensable for SsGH16 catalytic activity. Surface plasmon resonance experiments uncovered that both CBM32 domains specifically bind to αGal-epitope glycan, and together they had a KD of 3.5 mm toward a pentasaccharide αGal-epitope glycan. Cell-binding and αGal epitope removal assays revealed that SsGalNagA efficiently binds to both swine erythrocytes and tracheal epithelial cells and removes the αGal epitope from these cells, suggesting that SsGalNagA functions in nutrient acquisition or alters host signaling in S. suis Both binding and removal activities were blocked by an αGal-epitope glycan. SsGalNagA is the first enzyme reported to sequentially act on a glycan containing the αGal epitope. These findings shed detailed light on the evolution of GHs and an important host-pathogen interaction.
Collapse
Affiliation(s)
- Ping Chen
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Ran Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Mengmeng Huang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jinlu Zhu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Dong Wei
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Francis J Castellino
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA.,W. M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, Indiana, USA
| | - Guanghui Dang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Fang Xie
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Gang Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Ziyin Cui
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Siguo Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yueling Zhang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
31
|
Sasaki K, Harada M, Miyashita Y, Tagawa H, Kishimura A, Mori T, Katayama Y. Fc-binding antibody-recruiting molecules exploit endogenous antibodies for anti-tumor immune responses. Chem Sci 2020; 11:3208-3214. [PMID: 34122826 PMCID: PMC8157400 DOI: 10.1039/d0sc00017e] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Redirecting endogenous antibodies in the bloodstream to tumor cells using synthetic molecules is a promising approach to trigger anti-tumor immune responses. However, current molecular designs only enable the use of a small fraction of endogenous antibodies, limiting the therapeutic potential. Here, we report Fc-binding antibody-recruiting molecules (Fc-ARMs) as the first example addressing this issue. Fc-ARMs are composed of an Fc-binding peptide and a targeting ligand, enabling the exploitation of endogenous antibodies through constant affinity to the Fc region of antibodies, whose sequence is conserved in contrast to the Fab region. We show that Fc-ARM targeting folate receptor-α (FR-α) redirects a clinically used antibody mixture to FR-α+ cancer cells, resulting in cancer cell lysis by natural killer cells in vitro. Fc-ARMs successfully interacted with antibodies in vivo and accumulated in tumors. Furthermore, Fc-ARMs recruited antibodies to suppress tumor growth in a mouse model. Thus, Fc-ARMs have the potential to be a novel class of cancer immunotherapeutic agents. Fc-binding antibody-recruiting molecules provide robust and sufficient opportunities to employ endogenous antibodies for anti-tumor immune responses.![]()
Collapse
Affiliation(s)
- Koichi Sasaki
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University Fukuoka 819-0395 Japan
| | - Minori Harada
- Graduate School of Systems Life Sciences, Kyushu University Fukuoka 819-0395 Japan
| | - Yoshiki Miyashita
- Graduate School of Systems Life Sciences, Kyushu University Fukuoka 819-0395 Japan
| | - Hiroshi Tagawa
- Graduate School of Systems Life Sciences, Kyushu University Fukuoka 819-0395 Japan
| | - Akihiro Kishimura
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University Fukuoka 819-0395 Japan .,Graduate School of Systems Life Sciences, Kyushu University Fukuoka 819-0395 Japan.,International Research Center for Molecular Systems, Kyushu University Fukuoka 819-0395 Japan
| | - Takeshi Mori
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University Fukuoka 819-0395 Japan .,Graduate School of Systems Life Sciences, Kyushu University Fukuoka 819-0395 Japan
| | - Yoshiki Katayama
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University Fukuoka 819-0395 Japan .,Graduate School of Systems Life Sciences, Kyushu University Fukuoka 819-0395 Japan.,International Research Center for Molecular Systems, Kyushu University Fukuoka 819-0395 Japan.,Department of Biomedical Engineering, Chung Yuan Christian University Taoyuan Taiwan
| |
Collapse
|
32
|
Bilodeau C, Goltsis O, Rogers IM, Post M. Limitations of recellularized biological scaffolds for human transplantation. J Tissue Eng Regen Med 2019; 14:521-538. [PMID: 31826325 DOI: 10.1002/term.3004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 12/15/2022]
Abstract
A shortage of donor organs for transplantation and the dependence of the recipients on immunosuppressive therapy have motivated researchers to consider alternative regenerative approaches. The answer may reside in acellular scaffolds generated from cadaveric human and animal tissues. Acellular scaffolds are expected to preserve the architectural and mechanical properties of the original organ, permitting cell attachment, growth, and differentiation. Although theoretically, the use of acellular scaffolds for transplantation should pose no threat to the recipient's immune system, experimental data have revealed significant immune responses to allogeneic and xenogeneic transplanted scaffolds. Herein, we review the various factors of the scaffold that could trigger an inflammatory and/or immune response, thereby compromising its use for human transplant therapy. In addition, we provide an overview of the major cell types that have been considered for recellularization of the scaffold and their potential contribution to triggering an immune response.
Collapse
Affiliation(s)
- Claudia Bilodeau
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Olivia Goltsis
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Ian M Rogers
- Lunenfeld Research Institute, Mount Sinai Health, Toronto, Ontario, Canada
| | - Martin Post
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
33
|
|
34
|
Platt JL, Cascalho M, Piedrahita JA. Xenotransplantation: Progress Along Paths Uncertain from Models to Application. ILAR J 2019; 59:286-308. [PMID: 30541147 DOI: 10.1093/ilar/ily015] [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: 11/15/2017] [Revised: 08/23/2018] [Indexed: 12/18/2022] Open
Abstract
For more than a century, transplantation of tissues and organs from animals into man, xenotransplantation, has been viewed as a potential way to treat disease. Ironically, interest in xenotransplantation was fueled especially by successful application of allotransplantation, that is, transplantation of human tissue and organs, as a treatment for a variety of diseases, especially organ failure because scarcity of human tissues limited allotransplantation to a fraction of those who could benefit. In principle, use of animals such as pigs as a source of transplants would allow transplantation to exert a vastly greater impact than allotransplantation on medicine and public health. However, biological barriers to xenotransplantation, including immunity of the recipient, incompatibility of biological systems, and transmission of novel infectious agents, are believed to exceed the barriers to allotransplantation and presently to hinder clinical applications. One way potentially to address the barriers to xenotransplantation is by genetic engineering animal sources. The last 2 decades have brought progressive advances in approaches that can be applied to genetic modification of large animals. Application of these approaches to genetic engineering of pigs has contributed to dramatic improvement in the outcome of experimental xenografts in nonhuman primates and have encouraged the development of a new type of xenograft, a reverse xenograft, in which human stem cells are introduced into pigs under conditions that support differentiation and expansion into functional tissues and potentially organs. These advances make it appropriate to consider the potential limitation of genetic engineering and of current models for advancing the clinical applications of xenotransplantation and reverse xenotransplantation.
Collapse
Affiliation(s)
- Jeffrey L Platt
- Surgery, Microbiology & Immunology, and Transplantation Biology, University of Michigan, Ann Arbor, Michigan
| | - Marilia Cascalho
- Surgery, Microbiology & Immunology, and Transplantation Biology, University of Michigan, Ann Arbor, Michigan
| | - Jorge A Piedrahita
- Translational Medicine and The Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
35
|
Pipperger L, Koske I, Wild N, Müllauer B, Krenn D, Stoiber H, Wollmann G, Kimpel J, von Laer D, Bánki Z. Xenoantigen-Dependent Complement-Mediated Neutralization of Lymphocytic Choriomeningitis Virus Glycoprotein-Pseudotyped Vesicular Stomatitis Virus in Human Serum. J Virol 2019; 93:e00567-19. [PMID: 31243134 PMCID: PMC6714799 DOI: 10.1128/jvi.00567-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 06/18/2019] [Indexed: 01/09/2023] Open
Abstract
Neutralization by antibodies and complement limits the effective dose and thus the therapeutic efficacy of oncolytic viruses after systemic application. We and others previously showed that pseudotyping of oncolytic rhabdoviruses such as maraba virus and vesicular stomatitis virus (VSV) with the lymphocytic choriomeningitis virus glycoprotein (LCMV-GP) results in only a weak induction of neutralizing antibodies. Moreover, LCMV-GP-pseudotyped VSV (VSV-GP) was significantly more stable in normal human serum (NHS) than VSV. Here, we demonstrate that depending on the cell line used for virus production, VSV-GP showed different complement sensitivities in nonimmune NHS. The NHS-mediated titer reduction of VSV-GP was dependent on activation of the classical complement pathway, mainly by natural IgM antibodies against xenoantigens such as galactose-α-(1,3)-galactose (α-Gal) or N-glycolylneuraminic acid (Neu5Gc) expressed on nonhuman production cell lines. VSV-GP produced on human cell lines was stable in NHS. However, VSV-GP generated in transduced human cells expressing α-Gal became sensitive to NHS. Furthermore, GP-specific antibodies induced complement-mediated neutralization of VSV-GP independently of the producer cell line, suggesting that complement regulatory proteins potentially acquired by the virus during the budding process are not sufficient to rescue the virus from antibody-dependent complement-mediated lysis. Thus, our study points to the importance of a careful selection of cell lines for viral vector production for clinical use.IMPORTANCE Systemic application aims to deliver oncolytic viruses to tumors as well as to metastatic lesions. However, we found that xenoantigens incorporated onto the viral surface from nonhuman production cell lines are recognized by natural antibodies in human serum and that the virus is thereby inactivated by complement lysis. Hence, to maximize the effective dose, careful selection of cell lines for virus production is crucial.
Collapse
Affiliation(s)
- Lisa Pipperger
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Iris Koske
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Nicole Wild
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Brigitte Müllauer
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniela Krenn
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Heribert Stoiber
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Guido Wollmann
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, Innsbruck, Austria
| | - Janine Kimpel
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Dorothee von Laer
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Zoltán Bánki
- Division of Virology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
36
|
Mo F, Xue D, Duan S, Liu A, Yang X, Hou X, Lu X. Novel fusion cells derived from tumor cells expressing the heterologous α-galactose epitope and dendritic cells effectively target cancer. Vaccine 2019; 37:926-936. [PMID: 30661833 DOI: 10.1016/j.vaccine.2019.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 11/17/2018] [Accepted: 01/04/2019] [Indexed: 11/18/2022]
Abstract
Tumor cells/dendritic cells (DCs) fusion cells (tumor/DC) represent a promising immunotherapeutic strategy but are still under performed in clinical trials for cancer treatment. To further boost their anticancer efficacy, here we developed a novel design for fusing dendritic cells with MDA-MB-231 cells expressing the heterologous α-galactose (α-gal) epitope and assessed its anticancer activities both in vitro and in vivo. The high expression of α-gal in MDA-MB-231 (Gal+)/DC correlated with enhanced DC activation. When applied to T cells, MDA-MB-231 (Gal+)/DC significantly stimulated T-cell proliferation and activation, promoted productions of cytokines IL-2 and IFN-γ, and enhanced T-cell-mediated cytotoxicity against MDA-MB-231 cells. MDA-MB-231 (Gal+)/DC inhibited proliferation and promoted apoptosis of tumor cells in vivo, prolonged mouse survival, and significantly boosted anticancer immunity by increasing CD4+ and CD8+ T cells systemically and elevating serum levels of cytokines and IgG. These results suggested that fusing dendritic cells with tumor cells expressing the heterologous α-gal epitope provides a novel therapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
- Fengzhen Mo
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China; Intenational Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi 530021, China; Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Dabing Xue
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China; Intenational Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Siliang Duan
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China; Intenational Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Aiqun Liu
- Intenational Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xiaomei Yang
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China; Intenational Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xiaoqiong Hou
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China; Intenational Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xiaoling Lu
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China; Intenational Nanobody Research Center of Guangxi, Guangxi Medical University, Nanning, Guangxi 530021, China; National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China.
| |
Collapse
|
37
|
Hilger C, Fischer J, Wölbing F, Biedermann T. Role and Mechanism of Galactose-Alpha-1,3-Galactose in the Elicitation of Delayed Anaphylactic Reactions to Red Meat. Curr Allergy Asthma Rep 2019; 19:3. [PMID: 30673913 PMCID: PMC6344609 DOI: 10.1007/s11882-019-0835-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Purpose of Review The alpha-Gal (α-Gal) syndrome is characterized by the presence of IgE antibodies directed at the carbohydrate galactose-alpha-1,3-galactose (α-Gal). In this article, we review the presence of α-Gal in food and non-food sources; we discuss the evolutionary context of the antibody response to α-Gal and highlight immune responses to α-Gal and other carbohydrates. Recent findings IgE antibodies have been associated with delayed allergy to red meat. In addition to food, drugs, and other products of animal origin are increasingly perceived as a risk for patients sensitized to α-Gal. The link between tick bites and anti-α-Gal IgE-antibody production that has been established first by epidemiological studies has now been confirmed in mouse models. Summary The anti-α-Gal immune response is complex and characterized by a unique feature. IgM and IgG antibodies have been found to confer protection against pathogens whereas the IgE-response to α-Gal is detrimental and causes severe reactions upon exposure to mammalian meat and other products.
Collapse
Affiliation(s)
- Christiane Hilger
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg.
| | - Jörg Fischer
- Department of Dermatology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Florian Wölbing
- Department of Dermatology and Allergy Biederstein, Technical University of Munich, Munich, Germany
| | - Tilo Biedermann
- Department of Dermatology and Allergy Biederstein, Technical University of Munich, Munich, Germany.,Clinical Unit Allergology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| |
Collapse
|
38
|
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
|
39
|
Stahl EC, Bonvillain RW, Skillen CD, Burger BL, Hara H, Lee W, Trygg CB, Didier PJ, Grasperge BF, Pashos NC, Bunnell BA, Bianchi J, Ayares DL, Guthrie KI, Brown BN, Petersen TH. Evaluation of the host immune response to decellularized lung scaffolds derived from α-Gal knockout pigs in a non-human primate model. Biomaterials 2018; 187:93-104. [DOI: 10.1016/j.biomaterials.2018.09.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/21/2018] [Accepted: 09/23/2018] [Indexed: 12/11/2022]
|
40
|
Gal epitope expression and immunological properties in iGb3S deficient mice. Sci Rep 2018; 8:15433. [PMID: 30337628 PMCID: PMC6194060 DOI: 10.1038/s41598-018-33032-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 07/23/2018] [Indexed: 01/02/2023] Open
Abstract
The Gal antigen is synthesized by glycoprotein galactosyltransferase alpha 1, 3 (GGTA1) or (and) isoglobotrihexosylceramide 3 synthase (iGb3S). However, whether iGb3S deletion changes Gal epitope expression and immunological properties in animals is still not clear. The objective of this study was to develop iGb3S deficient mice, and characterize their Gal epitope expression and Gal epitope-related immunological properties. iGb3S gene knockout mice were generated on the C57BL/6 background using the bacterial artificial chromosome homology region recombination technique. Gal epitope expression in the iGb3S deficient mice was determined by using a monoclonal anti-Gal antibody. Immunological properties were analyzed by enzyme linked immune sorbent assay. It was found that Gal epitope expression was decreased from 5.19% to 21.74% in the main organs of iGb3S deficient mice, compared with that of C57BL/6 wild type mice, suggesting that the iGb3S gene participated to Gal epitope expression. However, iGb3S deletion alone did not cause significant changes in the immunological properties of iGb3S deficient mice with or without exogenous Gal antigen (Rabbit Red Blood Cell) stimulation. The data from this study suggest that the iGb3S gene likely contributes to Gal epitope expression, but may have a very weak effect on immunological properties of the iGb3S deficient mice.
Collapse
|
41
|
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
|
42
|
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
|
43
|
Younes M, Aggett P, Aguilar F, Crebelli R, Filipič M, Frutos MJ, Galtier P, Gott D, Gundert-Remy U, Kuhnle GG, Lambré C, Leblanc JC, Lillegaard IT, Moldeus P, Mortensen A, Oskarsson A, Stankovic I, Waalkens-Berendsen I, Woutersen RA, Wright M, Brimer L, Lindtner O, Mosesso P, Christodoulidou A, Ioannidou S, Lodi F, Dusemund B. Re-evaluation of carrageenan (E 407) and processed Eucheuma seaweed (E 407a) as food additives. EFSA J 2018; 16:e05238. [PMID: 32625873 PMCID: PMC7009739 DOI: 10.2903/j.efsa.2018.5238] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The present opinion deals with the re-evaluation of the safety of food-grade carrageenan (E 407) and processes Eucheuma seaweed (E 407a) used as food additives. Because of the structural similarities, the Panel concluded that processed Eucheuma seaweed can be included in the evaluation of food-grade carrageenan. Poligeenan (average molecular weight 10-20 kDa) has not been authorised as a food additive and is not used in any food applications. In its evaluation of carrageenan (E 407) and processed Eucheuma seaweed (E 407a), the Panel noted that the ADME database was sufficient to conclude that carrageenan was not absorbed intact; in a subchronic toxicity study performed with carrageenan almost complying with the EU specification for E 407 in rats, the no-observed-adverse-effect level (NOAEL) was 3,400-3,900 mg/kg body weight (bw) per day, the highest dose tested; no adverse effects have been detected in chronic toxicity studies with carrageenan in rats up to 7,500 mg/kg bw per day, the highest dose tested; there was no concern with respect to the carcinogenicity of carrageenan; carrageenan and processed Eucheuma seaweed did not raise a concern with respect to genotoxicity; the NOAEL of sodium and calcium carrageenan for prenatal developmental dietary toxicity studies were the highest dose tested; the safety of processed Eucheuma seaweed was sufficiently covered by the toxicological evaluation of carrageenan; data were adequate for a refined exposure assessment for 41 out of 79 food categories. However, the Panel noted uncertainties as regards the chemistry, the exposure assessment and biological and toxicological data. Overall, taking into account the lack of adequate data to address these uncertainties, the Panel concluded that the existing group acceptable daily intake (ADI) for carrageenan (E 407) and processed Eucheuma seaweed (E 407a) of 75 mg/kg bw per day should be considered temporary, while the database should be improved within 5 years after publication of this opinion.
Collapse
|
44
|
Anraku K, Sato S, Jacob NT, Eubanks LM, Ellis BA, Janda KD. The design and synthesis of an α-Gal trisaccharide epitope that provides a highly specific anti-Gal immune response. Org Biomol Chem 2018; 15:2979-2992. [PMID: 28294277 DOI: 10.1039/c7ob00448f] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Carbohydrate antigens displaying Galα(1,3)Gal epitopes are recognized by naturally occurring antibodies in humans. These anti-Gal antibodies comprise up to 1% of serum IgG and have been viewed as detrimental as they are responsible for hyperacute organ rejections. In order to model this condition, α(1,3)galactosyltransferase-knockout mice are inoculated against the Galα(1,3)Gal epitope. In our study, two α-Gal trisaccharide epitopes composed of either Galα(1,3)Galβ(1,4)GlcNAc or Galα(1,3)Galβ(1,4)Glc linked to a squaric acid ester moiety were examined for their ability to elicit immune responses in KO mice. Both target epitopes were synthesized using a two-component enzymatic system using modified disaccharide substrates containing a linker moiety for coupling. While both glycoconjugate vaccines induced the required high anti-Gal IgG antibody titers, it was found that this response had exquisite specificity for the Galα(1,3)Galβ(1,4)GlcNAc hapten used, with little cross reactivity with the Galα(1,3)Galβ(1,4)Glc hapten. Our findings indicate that while homogenous glycoconjugate vaccines provide high IgG titers, the carrier and adjuvanting factors can deviate the specificity to an antigenic determinant outside the purview of interest.
Collapse
Affiliation(s)
- Kensaku Anraku
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, The Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, 10550 N Torrey Pines Rd BCC-582, La Jolla, CA 92037, USA. and Department of Medical Technology, Kumamoto Health Science University, 325 Izumi-machi, Kita-ku, Kumamoto 861-5598, Japan
| | - Shun Sato
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, The Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, 10550 N Torrey Pines Rd BCC-582, La Jolla, CA 92037, USA. and Research Institute for Sustainable Chemistry, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba Central 5-2, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Nicholas T Jacob
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, The Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, 10550 N Torrey Pines Rd BCC-582, La Jolla, CA 92037, USA.
| | - Lisa M Eubanks
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, The Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, 10550 N Torrey Pines Rd BCC-582, La Jolla, CA 92037, USA.
| | - Beverly A Ellis
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, The Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, 10550 N Torrey Pines Rd BCC-582, La Jolla, CA 92037, USA.
| | - Kim D Janda
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, The Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, 10550 N Torrey Pines Rd BCC-582, La Jolla, CA 92037, USA.
| |
Collapse
|
45
|
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
|
46
|
Cravedi P, Farouk S, Angeletti A, Edgar L, Tamburrini R, Duisit J, Perin L, Orlando G. Regenerative immunology: the immunological reaction to biomaterials. Transpl Int 2017; 30:1199-1208. [PMID: 28892571 DOI: 10.1111/tri.13068] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 06/29/2017] [Accepted: 09/04/2017] [Indexed: 01/09/2023]
Abstract
Regenerative medicine promises to meet two of the most urgent needs of modern organ transplantation, namely immunosuppression-free transplantation and an inexhaustible source of organs. Ideally, bioengineered organs would be manufactured from a patient's own biomaterials-both cells and the supporting scaffolding materials in which cells would be embedded and allowed to mature to eventually regenerate the organ in question. While some groups are focusing on the feasibility of this approach, few are focusing on the immunogenicity of the scaffolds that are being developed for organ bioengineering purposes. This review will succinctly discuss progress in the understanding of immunological characteristics and behavior of different scaffolds currently under development, with emphasis on the extracellular matrix scaffolds obtained decellularized animal or human organs which seem to provide the ideal template for bioengineering purposes.
Collapse
Affiliation(s)
- Paolo Cravedi
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samira Farouk
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrea Angeletti
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Experimental, Diagnostic, Specialty Medicine, Nephrology, Dialysis, and Renal Transplant Unit, S. Orsola University Hospital, Bologna, Italy
| | - Lauren Edgar
- Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Riccardo Tamburrini
- Wake Forest University School of Medicine, Winston Salem, NC, USA.,Section of Transplantation, Department of Surgery, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Jerome Duisit
- Pôle de Chirurgie Expérimentale (CHEX), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Plastic and Reconstructive Surgery, Cliniques Universitaires St-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Laura Perin
- Division of Urology, GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics, Saban Research Institute, Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Giuseppe Orlando
- Wake Forest University School of Medicine, Winston Salem, NC, USA.,Section of Transplantation, Department of Surgery, Wake Forest University School of Medicine, Winston Salem, NC, USA
| |
Collapse
|
47
|
Alpha-Gal Syndrome: Clinical Presentation, New Concepts, and Unmet Needs. CURRENT TREATMENT OPTIONS IN ALLERGY 2017. [DOI: 10.1007/s40521-017-0134-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
48
|
Abstract
BACKGROUND The rapidly improving tools of genetic engineering may make it possible to overcome the humoral immune barrier that prevents xenotransplantation. We hypothesize that levels of human antibody binding to donor tissues from swine must approximate the antibody binding occurring in allotransplantation. It is uncertain if this is an attainable goal. Here we perform an initial analysis of this issue by comparing human antibody binding to red blood cells (RBC) isolated from knockout swine and to allogeneic or autologous human RBC. METHODS Human sera were incubated with RBC isolated from various genetically engineered swine or from humans. The level of IgG and IgM binding to these cells were compared using either flow cytometry or a novel mass spectrometric assay. RESULTS Mass spectroscopic quantitation of human antibody binding demonstrated that as few as 3 gene inactivations can reduce the levels human antibody binding to swine RBC that is as low as autologous human RBC. Flow cytometry showed that RBC from 2-gene knockout swine exhibited less human antibody binding than human blood group O allogeneic RBC in 22% of tested sera. Deletion of a third gene from pigs resulted in 30% of human samples having less IgG and IgM RBC xenoreactivity than alloreactivity. CONCLUSIONS Xenoantigenicity of swine RBC can be eliminated via gene disruption. These results suggest that the gene knockout approach may be able reduce antigenicity in other pig tissues to levels that enable the xenotransplantation humoral barrier to be overcome.
Collapse
|
49
|
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
|
50
|
Stone KR, Walgenbach A, Galili U. Induced Remodeling of Porcine Tendons to Human Anterior Cruciate Ligaments by α-GAL Epitope Removal and Partial Cross-Linking. TISSUE ENGINEERING PART B-REVIEWS 2017; 23:412-419. [PMID: 28068870 PMCID: PMC5567590 DOI: 10.1089/ten.teb.2016.0332] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
This review describes a novel method developed for processing porcine tendon and other ligament implants that enables in situ remodeling into autologous ligaments in humans. The method differs from methods using extracellular matrices (ECMs) that provide postoperative orthobiological support (i.e., augmentation grafts) for healing of injured ligaments, in that the porcine bone-patellar-tendon-bone itself serves as the graft replacing ruptured anterior cruciate ligament (ACL). The method allows for gradual remodeling of porcine tendon into autologous human ACL while maintaining the biomechanical integrity. The method was first evaluated in a preclinical model of monkeys and subsequently in patients. The method overcomes detrimental effects of the natural anti-Gal antibody and harnesses anti-non-gal antibodies for the remodeling process in two steps: Step 1. Elimination of α-gal epitopes—this epitope that is abundant in pigs (as in other nonprimate mammals) binds the natural anti-Gal antibody, which is the most abundant natural antibody in humans. This interaction, which can induce fast resorption of the porcine implant, is avoided by enzymatic elimination of α-gal epitopes from the implant with recombinant α-galactosidase. Step 2. Partial cross-linking of porcine tendon with glutaraldehyde—this cross-linking generates covalent bonds in the ECM, which slow infiltration of macrophages into the implant. Anti-non-gal antibodies are produced in recipients against the multiple porcine antigenic proteins and proteoglycans because of sequence differences between human and porcine homologous proteins. Anti-non-gal antibodies bind to the implant ECM, recruit macrophages, and induce the implant destruction by directing proteolytic activity of macrophages. Partial cross-linking of the tendon ECM decreases the extent of macrophage infiltration and degradation of the implant and enables concomitant infiltration of fibroblasts that follow the infiltrating macrophages. These fibroblasts align with the implant collagen fibers and secrete their own collagen and other ECM proteins, which gradually remodel the porcine tendon into human ACL. This ligamentization process lasts ∼2 years and the biomechanical integrity of the graft is maintained throughout the whole period. These studies are the first, and so far the only, to demonstrate remodeling of porcine tendon implants into permanently functional autologous ACL in humans.
Collapse
Affiliation(s)
- Kevin R Stone
- 1 The Stone Clinic and Foundation , San Francisco, California
| | - Ann Walgenbach
- 1 The Stone Clinic and Foundation , San Francisco, California
| | - Uri Galili
- 2 Department of Surgery, University of Massachusetts Medical School , Worcester, Massachusetts (retired)
| |
Collapse
|