Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters

Database
Language
Journal subject
Affiliation country
Publication year range
1.
Pathogens ; 11(5)2022 May 16.
Article in English | MEDLINE | ID: mdl-35631109

ABSTRACT

Immune cell invasion after the transplantation of solid organs is directed by chemokines binding to glycosaminoglycans (GAGs), creating gradients that guide immune cell infiltration. Renal transplant is the preferred treatment for end stage renal failure, but organ supply is limited and allografts are often injured during transport, surgery or by cytokine storm in deceased donors. While treatment for adaptive immune responses during rejection is excellent, treatment for early inflammatory damage is less effective. Viruses have developed highly active chemokine inhibitors as a means to evade host responses. The myxoma virus-derived M-T7 protein blocks chemokine: GAG binding. We have investigated M-T7 and also antisense (ASO) as pre-treatments to modify chemokine: GAG interactions to reduce donor organ damage. Immediate pre-treatment of donor kidneys with M-T7 to block chemokine: GAG binding significantly reduced the inflammation and scarring in subcapsular and subcutaneous allografts. Antisense to N-deacetylase N-sulfotransferase1 (ASONdst1) that modifies heparan sulfate, was less effective with immediate pre-treatment, but reduced scarring and C4d staining with donor pre-treatment for 7 days before transplantation. Grafts with conditional Ndst1 deficiency had reduced inflammation. Local inhibition of chemokine: GAG binding in donor organs immediately prior to transplant provides a new approach to reduce transplant damage and graft loss.

2.
Methods Mol Biol ; 2225: 257-273, 2021.
Article in English | MEDLINE | ID: mdl-33108668

ABSTRACT

Solid tissue transplant is a growing medical need that is further complicated by a limited donor organ supply. Acute and chronic rejection occurs in nearly all transplants and reduces long-term graft survival, thus increasing the need for repeat transplantation. Viruses have evolved highly adapted responses designed to evade the host's immune defenses. Immunomodulatory proteins derived from viruses represent a novel class of potential therapeutics that are under investigation as biologics to attenuate immune-mediated rejection and damage. These immune-modulating proteins have the potential to reduce the need for traditional posttransplant immune suppressants and improve graft survival. The myxoma virus-derived protein M-T7 is a promising biologic that targets chemokine and glycosaminoglycan pathways central to kidney transplant rejection. Orthotopic transplantations in mice are prohibitively difficult and costly and require a highly trained microsurgeon to successfully perform the procedure. Here we describe a kidney-to-kidney subcapsular transplant model as a practical and simple method for studying transplant rejection, a model that requires fewer mice. One kidney can be used as a donor for transplants into six or more recipient mice. Using this model there is lower morbidity, pain, and mortality for the mice. Subcapsular kidney transplantation provides a first step approach to testing virus-derived proteins as new potential immune-modulating therapeutics to reduce transplant rejection and inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Graft Rejection/prevention & control , Graft Survival , Immunologic Factors/pharmacology , Kidney Transplantation/methods , Myxoma virus/chemistry , Viral Proteins/pharmacology , Animals , Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/metabolism , Biomarkers/analysis , Chemokines/biosynthesis , Complement C4b/genetics , Complement C4b/immunology , Female , Gene Expression , Graft Rejection/genetics , Graft Rejection/immunology , Graft Rejection/pathology , Immunologic Factors/biosynthesis , Immunologic Factors/immunology , Kidney/immunology , Kidney/surgery , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptide Fragments/genetics , Peptide Fragments/immunology , Receptors, Interferon/biosynthesis , Receptors, Interferon/immunology , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Transplantation, Homologous , Viral Proteins/biosynthesis , Viral Proteins/immunology
3.
Methods Mol Biol ; 2225: 217-226, 2021.
Article in English | MEDLINE | ID: mdl-33108665

ABSTRACT

Immune modulators play critical roles in the progression of wounds to normal or conversely delayed healing, through the regulation of normal tissue regrowth, scarring, inflammation, and growth factor expression. Many immune modulator recombinants are under active preclinical study or in clinical trial to promote improved acute or chronic wound healing and to reduce scarring. Viruses have evolved highly efficient immune modulators for the evasion of host-defensive immune responses that target and kill invasive viruses. Recent studies have proven that some of these virus-derived immune modulators can be used to promote wound healing with significantly improved speed and reduced scarring in rodent models. Mouse full-thickness excisional wound model is one of the most commonly used animal models used to study wound healing for its similarity to humans in the healing phases and associated cellular and molecular mechanisms. This chapter introduces this mouse dermal wound healing model in detail for application in studying viral immune modulators as new treatments to promote wound healing. Details of hydrogel, protein construction, and topical application methods for these therapeutic proteins are provided in this chapter.


Subject(s)
Cicatrix/prevention & control , Immunologic Factors/pharmacology , Myxoma virus/chemistry , Surgical Wound/drug therapy , Viral Proteins/pharmacology , Wound Healing/drug effects , Administration, Cutaneous , Animals , Chitosan/chemistry , Cicatrix/genetics , Cicatrix/immunology , Cicatrix/pathology , Collagen Type I/biosynthesis , Collagen Type I/genetics , Disease Models, Animal , Drug Delivery Systems , Female , Gene Expression , Hydrogels/chemistry , Immunologic Factors/immunology , Male , Mice , Mice, Inbred C57BL , Skin/drug effects , Skin/injuries , Surgical Wound/genetics , Surgical Wound/immunology , Surgical Wound/pathology , Viral Proteins/immunology , Wound Healing/immunology
4.
Pharmaceutics ; 12(11)2020 Oct 22.
Article in English | MEDLINE | ID: mdl-33105865

ABSTRACT

Complex dermal wounds represent major medical and financial burdens, especially in the context of comorbidities such as diabetes, infection and advanced age. New approaches to accelerate and improve, or "fine tune" the healing process, so as to improve the quality of cutaneous wound healing and management, are the focus of intense investigation. Here, we investigate the topical application of a recombinant immune modulating protein which inhibits the interactions of chemokines with glycosaminoglycans, reducing damaging or excess inflammation responses in a splinted full-thickness excisional wound model in mice. M-T7 is a 37 kDa-secreted, virus-derived glycoprotein that has demonstrated therapeutic efficacy in numerous animal models of inflammatory immunopathology. Topical treatment with recombinant M-T7 significantly accelerated wound healing when compared to saline treatment alone. Healed wounds exhibited properties of improved tissue remodeling, as determined by collagen maturation. M-T7 treatment accelerated the rate of peri-wound angiogenesis in the healing wounds with increased levels of TNF, VEGF and CD31. The immune cell response after M-T7 treatment was associated with a retention of CCL2 levels, and increased abundances of arginase-1-expressing M2 macrophages and CD4 T cells. Thus, topical treatment with recombinant M-T7 promotes a pro-resolution environment in healing wounds, and has potential as a novel treatment approach for cutaneous tissue repair.

SELECTION OF CITATIONS
SEARCH DETAIL