Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 354
Filter
2.
Drug Deliv ; 29(1): 925-936, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35311607

ABSTRACT

Antigen-specific immune tolerance, which possesses great potential in preventing or curing type 1 diabetes mellitus (T1DM), can be induced by oral vaccination with T1DM-related autoantigens. However, direct administration of autoantigens via oral route exhibits a low tolerance-inducing effect as a result of the digestion of protein antigens in the gastrointestinal tract (GIT) and therefore, a large dosage of autoantigens may be needed. In this study, bacterium-like particles (BLPs) made from food-grade lactic acid bacteria were used to deliver the intracellular domain of the insulinoma-associated protein 2 (IA-2ic). For this purpose, BLPs-IA-2ic vaccine in which IA-2ic bound to the surface of BLPs was constructed. BLPs enhanced the stability of the delivered IA-2ic based on the stability analysis in vitro. Oral administration of BLPs-IA-2ic significantly reduced T1DM incidence in NOD mice. The mice fed BLPs-IA-2ic exhibited a significant reduction in insulitis and preserved the ability to secrete insulin. Immunologic analysis showed that oral vaccination with BLPs-IA-2ic induced antigen-specific T cell tolerance. The results revealed that the successful induction of immune tolerance was dependent on the immune deviation (in favor of T helper 2 responses) and CD4+CD25+FoxP3+ regulatory T cells. Hence, oral vaccination with BLPs-IA-2ic shows potential for application in preventing T1DM.


Subject(s)
Autoantigens , Diabetes Mellitus, Type 1 , Receptor-Like Protein Tyrosine Phosphatases, Class 8 , Animals , Autoantigens/administration & dosage , Diabetes Mellitus, Type 1/prevention & control , Insulin , Mice , Mice, Inbred NOD , Receptor-Like Protein Tyrosine Phosphatases, Class 8/administration & dosage , T-Lymphocytes, Regulatory
3.
Mol Pharm ; 18(4): 1806-1818, 2021 04 05.
Article in English | MEDLINE | ID: mdl-33734705

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune subtype of diabetes, mainly caused by the immune attack of self-insulin-producing cells. Immune modulation that delays the onset of T1D is able to reduce diabetic complications and mortality. We have previously reported that mannosylated sodium alginate nanoparticles (MAN-ALG) exhibited excellent dendritic cell targeting and in vivo antigen delivery efficacy. To investigate the role of MAN-ALG in an autoimmune context, we loaded the MAN-ALG with Ins29-23, a T1D autoantigen [MAN-ALG(PEP)], for T1D immune tolerance induction in nonobese diabetic (NOD) mice. We observed the delayed onset of T1D occurrence and some degree of blood glucose reduction accompanied by a larger islet area, attributable to augmented T-regulatory cell proportion in mice treated with MAN-ALG(PEP). However, MAN-ALG was also found to elicit lysosomal escape and cross-presentation of Ins29-23 in bone marrow-derived dendritic cells, leading to the immune activation of Ins29-23-recognizing T cells and destruction of Ins29-23-expressing islet cells. This dual impact resulted in delayed but a nonpreventive effect of MAN-ALG(PEP) on the T1D onset in NOD mice. Considering the potent immune stimulatory property of MAN-ALG, cautions should be implemented when using alginate-based biomaterials in an autoimmune context. Moreover, it is also noted that regarding the in vivo outcome of immune therapies, biomaterial-based delivery systems and their detailed role on immune regulation need to be examined.


Subject(s)
Autoantigens/administration & dosage , Diabetes Mellitus, Type 1/prevention & control , Drug Carriers/chemistry , Insulin/immunology , Peptides/administration & dosage , Alginates/chemistry , Animals , Autoantigens/genetics , Autoantigens/immunology , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Female , Humans , Immune Tolerance , Insulin/genetics , Islets of Langerhans/immunology , Mice , Mice, Inbred NOD , Nanoparticles/chemistry , Peptides/genetics , Peptides/immunology
4.
Immunology ; 162(4): 452-463, 2021 04.
Article in English | MEDLINE | ID: mdl-33346377

ABSTRACT

Autoimmune diseases are caused by adaptive immune responses to self-antigens. The development of antigen-specific therapies that suppress disease-related, but not unrelated immune responses in general, is an important goal of biomedical research. We have previously shown that delivery of myelin peptides to liver sinusoidal endothelial cells (LSECs) using LSEC-targeting nanoparticles provides effective protection from CD4 T-cell-driven autoimmune encephalomyelitis. Here, we investigated whether this methodology might also serve antigen-specific treatment of a CD8 T-cell-driven autoimmune disease. As a model for CD8 T-cell-mediated autoimmunity, we used OT-1 T-cell-driven cholangitis in K14-OVAp mice expressing the cognate MHC I-restricted SIINFEKL peptide in cholangiocytes. To study whether peptide delivery to LSECs could modulate cholangitis, SIINFEKL peptide-conjugated nanoparticles were administered intravenously one day before transfer of OT-1 T cells; five days after cell transfer, liver pathology and hepatic infiltrates were analysed. SIINFEKL peptide-conjugated nanoparticles were rapidly taken up by LSECs in vivo, which effectively cross-presented the delivered peptide on MHC I molecules. Intriguingly, K14-OVAp mice receiving SIINFEKL-loaded nanoparticles manifested significantly reduced liver damage compared with vehicle-treated K14-OVAp mice. Mechanistically, treatment with LSEC-targeting SIINFEKL-loaded nanoparticles significantly reduced the number of liver-infiltrating OT-1 T cells, which up-regulated expression of the co-inhibitory receptor PD-1 and down-regulated cytotoxic effector function and inflammatory cytokine production. These findings show that tolerogenic LSECs can effectively internalize circulating nanoparticles and cross-present nanoparticle-bound peptides on MHC I molecules. Therefore, nanoparticle-mediated autoantigen peptide delivery to LSECs might serve the antigen-specific treatment of CD8 T-cell-driven autoimmune disease.


Subject(s)
Autoantigens/administration & dosage , Autoimmune Diseases/immunology , CD8-Positive T-Lymphocytes/immunology , Cholangitis/immunology , Endothelial Cells/immunology , Immunotherapy/methods , Liver/pathology , Magnetic Iron Oxide Nanoparticles/administration & dosage , Ovalbumin/administration & dosage , T-Lymphocytes, Regulatory/immunology , Animals , Autoantigens/chemistry , Autoimmune Diseases/therapy , Cells, Cultured , Cholangitis/therapy , Cross-Priming , Cytotoxicity, Immunologic , Disease Models, Animal , Humans , Immunosuppression Therapy , Liver/blood supply , Magnetic Iron Oxide Nanoparticles/chemistry , Mice , Mice, Transgenic , Ovalbumin/chemistry , Peptide Fragments/administration & dosage , Peptide Fragments/chemistry , Programmed Cell Death 1 Receptor/metabolism
6.
Mol Pharm ; 17(11): 4201-4211, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32903002

ABSTRACT

Many autoimmune therapies focus on immune suppression to reduce symptom severity and halt disease progression; however, currently approved treatments lack specificity for the autoantigen and rely on more global immune suppression. Multivalent antigen arrays can disarm pathogenic autoimmune B cell populations that specifically recognize the antigen of interest via their B cell receptor (BCR). Disarmament may be achieved by BCR engagement, cross-linking, and sustained receptor occupancy as a result of multivalent, high avidity BCR binding. To engage and explore this mechanism, a tetramer display of the encephalogenic proteolipid peptide (PLP139-151), referred to as 4-arm PLP139-151, was synthesized by copper-catalyzed azide-alkyne cycloaddition chemistry. Subcutaneous administration of 4-arm PLP139-151 completely ameliorated symptoms of paralysis in a mouse model of multiple sclerosis known as experimental autoimmune encephalomyelitis. Competitive binding of 4-arm PLP139-151 to PLP139-151-specific IgG in the mouse serum demonstrated the enhanced avidity associated with the multivalent array compared to the free peptide. Furthermore, key PLP139-151-reactive B cells were depleted following 4-arm PLP139-151 treatment, resulting in significant reduction of proinflammatory cytokines. Together, these data demonstrate the potential of 4-arm PLP139-151 to silence autoreactive B cell populations and limit the downstream activation of effector cells.


Subject(s)
Autoantigens/administration & dosage , B-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Immune Tolerance , Immunotherapy/methods , Multiple Sclerosis/therapy , Myelin Proteolipid Protein/administration & dosage , Peptide Fragments/administration & dosage , Administration, Topical , Animals , Autoantigens/blood , Autoantigens/immunology , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Immunoglobulin G/blood , Mice , Multiple Sclerosis/blood , Multiple Sclerosis/immunology , Myelin Proteolipid Protein/blood , Myelin Proteolipid Protein/immunology , Paralysis/blood , Paralysis/immunology , Paralysis/therapy , Peptide Fragments/blood , Peptide Fragments/immunology , Receptors, Antigen, B-Cell/immunology , Treatment Outcome
7.
Cytokine ; 136: 155227, 2020 12.
Article in English | MEDLINE | ID: mdl-32810784

ABSTRACT

Sepsis is the most common critical disease with high mortality in intensive care unit. Platelet count (PC) frequently altered in sepsis patients and implicated in the pathogenesis of multi-organ failure. It is also worth mentioning that thrombocytopenia was closely associated with poor outcomes in sepsis patients. However, whether drug intervention aimed at correcting thrombocytopenia would improve the prognosis of sepsis patients and which kind of sepsis patients could benefit from this therapy is still unclear. This study aims to explore the effect of severe thrombocytopenia on the prognosis of sepsis and the impact of a platelet-elevating drug (recombinant human thrombopoietin, rhTPO) for these sepsis patients. In this study, we included 249 sepsis patients diagnosed by sepsis 3.0, and these patients were classified into the three groups based on PC: normal (PC ≥ 100 × 109/L), mild-moderate thrombocytopenia (50 × 109/L ≤ PC < 100 × 109/L), and severe thrombocytopenia (PC < 50 × 109/L). We found that patients with severe thrombocytopenia had more blood transfusion, shorter days free from organ support, and worse outcomes as compared with the normal group. However, there was no significant difference between normal and mild-moderate thrombocytopenia groups. Furthermore, a subgroup analysis showed that rescue therapy with rhTPO could rapidly lead to a recovery of the PC, prolong days free from organ support, increase survival days, and reduce the 28-day mortality in sepsis patients with severe thrombocytopenia. These results suggested that sepsis patients with severe thrombocytopenia, not mild-moderate thrombocytopenia, had a poorer prognosis. RhTPO, probably as effective rescue therapy, could quickly recover PC and improve the prognosis in these sepsis patients.


Subject(s)
Autoantigens/administration & dosage , Blood Transfusion , Iodide Peroxidase/administration & dosage , Iron-Binding Proteins/administration & dosage , Sepsis , Thrombocytopenia , Aged , Critical Illness , Disease-Free Survival , Female , Humans , Male , Middle Aged , Recombinant Proteins/administration & dosage , Sepsis/blood , Sepsis/complications , Sepsis/mortality , Sepsis/therapy , Survival Rate , Thrombocytopenia/blood , Thrombocytopenia/etiology , Thrombocytopenia/mortality , Thrombocytopenia/therapy
8.
Clin Exp Med ; 20(3): 381-385, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32458092

ABSTRACT

To review the efficacy and safety of glucocorticoids combined with different regimens for treating severe immune thrombocytopenia (ITP). Eighty-five severe ITP patients from 2 tertiary hospitals treated with glucocorticoids were enrolled from January 2018 to May 2019 and divided into 4 treatment groups: group A (treated with glucocorticoids), group B (glucocorticoids plus intravenous immunoglobulin (IVIg)), group C (glucocorticoids plus recombinant human thrombopoietin (rhTPO)), and group D (glucocorticoids plus IVIg and rhTPO). Statistical analysis was performed with SPSS 19.0 software. Early responses and response maintenance were assessed at 14 days and 1 month after treatment. Groups B, C and D had higher complete response (CR) and overall response (OR) rates than group A (P < 0.05). Adverse reaction incidences were not significantly different among all groups (P > 0.05). Severe ITP patients who received glucocorticoids with IVIg and rhTPO had higher CRs and ORs at the platelet level, and no significant adverse reactions were observed. Glucocorticoids combined with different regimens had different clinical efficacies for treating severe ITP.


Subject(s)
Autoantigens/administration & dosage , Glucocorticoids/administration & dosage , Immunoglobulins, Intravenous/administration & dosage , Iodide Peroxidase/administration & dosage , Iron-Binding Proteins/administration & dosage , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Adult , Aged , Autoantigens/adverse effects , China , Drug Administration Schedule , Drug Therapy, Combination , Female , Glucocorticoids/adverse effects , Humans , Immunoglobulins, Intravenous/adverse effects , Iodide Peroxidase/adverse effects , Iron-Binding Proteins/adverse effects , Male , Middle Aged , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Retrospective Studies , Treatment Outcome , Young Adult
9.
Int J Mol Sci ; 21(5)2020 Feb 26.
Article in English | MEDLINE | ID: mdl-32111075

ABSTRACT

Autoantigen treatment has been tried for the prevention of type 1 diabetes (T1D) and to preserve residual beta-cell function in patients with a recent onset of the disease. In experimental animal models, efficacy was good, but was insufficient in human subjects. Besides the possible minor efficacy of peroral insulin in high-risk individuals to prevent T1D, autoantigen prevention trials have failed. Other studies on autoantigen prevention and intervention at diagnosis are ongoing. One problem is to select autoantigen/s; others are dose and route. Oral administration may be improved by using different vehicles. Proinsulin peptide therapy in patients with T1D has shown possible minor efficacy. In patients with newly diagnosed T1D, subcutaneous injection of glutamic acid decarboxylase (GAD) bound to alum hydroxide (GAD-alum) can likely preserve beta-cell function, but the therapeutic effect needs to be improved. Intra-lymphatic administration may be a better alternative than subcutaneous administration, and combination therapy might improve efficacy. This review elucidates some actual problems of autoantigen therapy in the prevention and/or early intervention of type 1 diabetes.


Subject(s)
Autoantigens/administration & dosage , Autoantigens/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Administration, Oral , Animals , Autoantigens/immunology , Chemotherapy, Adjuvant , Drug Therapy, Combination , Glutamate Decarboxylase/therapeutic use , Humans , Injections, Intralymphatic , Injections, Subcutaneous , Insulin/metabolism , Proinsulin/therapeutic use , Vitamin D/therapeutic use
10.
Am J Nephrol ; 51(2): 99-107, 2020.
Article in English | MEDLINE | ID: mdl-31927545

ABSTRACT

BACKGROUND AND AIM: Primary membranous nephropathy (pMN) is the most common cause of nephrotic syndrome in adults. Recent studies suggested that immunization of DBA/1 mice with the main antigen of antiglomerular basement membrane disease (GBM) disease, α3(IV)NC1, could lead to MN lesions. This study aimed to explore the pathogenic epitopes for mouse MN. METHODS: Twenty-four linear peptides were synthesized spanning human α3(IV)NC1. Male DBA/1 mice aged 6-8 weeks were immunized with the peptides 200 µg/mouse in Freund's complete adjuvant subcutaneously and boosted 3 times with the peptides in Freund's incomplete adjuvant in weeks 3, 5, and 7. The blood and 24-h urine samples were assessed every 2 weeks. The kidneys were examined when the mice were sacrificed at 18 weeks. RESULTS: All the mice immunized with human α3(IV)NC1 and the 24 peptides produced circulating antibodies against the immunogens at 2 weeks and achieved the maximum titers at 8 weeks. About 5/6 (83%) mice immunized with α3(IV)NC1 and (3/6) 50% of the mice immunized with peptide 23 (α3141-154) showed proteinuria at 8-10 weeks and increased continuously. The kidneys showed granular depositions of IgG, C3, and C5b-9 along the glomerular capillary loops. The major IgG subclass was IgG1 (equivalent to human IgG4). GBM thickening with the formation of spikes and subepithelial electron-dense deposits were observed under electron microscope. CONCLUSION: The linear peptide of α3141-154 could induce clinical and histopathological features of MN in DBA/1 mice, which might give clues to the mechanism of MN in combination with anti-GBM disease.


Subject(s)
Autoantigens/administration & dosage , Collagen Type IV/administration & dosage , Epitope Mapping , Glomerulonephritis, Membranous/immunology , Immunization , Animals , Male , Mice
11.
Front Immunol ; 11: 606936, 2020.
Article in English | MEDLINE | ID: mdl-33613527

ABSTRACT

Systemic lupus erythematosus (SLE) and other autoimmune diseases are propelled by immune dysregulation and pathogenic, disease-specific autoantibodies. Autoimmunity against the lupus autoantigen Sm is associated with cross-reactivity to Epstein-Barr virus (EBV) nuclear antigen 1 (EBNA-1). Additionally, EBV latent membrane protein-1 (LMP1), initially noted for its oncogenic activity, is an aberrantly active functional mimic of the B cell co-stimulatory molecule CD40. Mice expressing a transgene (Tg) for the mCD40-LMP1 hybrid molecule (containing the cytoplasmic tail of LMP1) have mild autoantibody production and other features of immune dysregulation by 2-3 months of age, but no overt autoimmune disease. This study evaluates whether exposure to the EBV molecular mimic, EBNA-1, stimulates antigen-specific and concurrently-reactive humoral and cellular immunity, as well as lupus-like features. After immunization with EBNA-1, mCD40-LMP1 Tg mice exhibited enhanced, antigen-specific, cellular and humoral responses compared to immunized WT congenic mice. EBNA-1 specific proliferative and inflammatory cytokine responses, including IL-17 and IFN-γ, were significantly increased (p<0.0001) in mCD40-LMP1 Tg mice, as well as antibody responses to amino- and carboxy-domains of EBNA-1. Of particular interest was the ability of mCD40-LMP1 to drive EBNA-1 associated molecular mimicry with the lupus-associated autoantigen, Sm. EBNA-1 immunized mCD40-LMP1 Tg mice exhibited enhanced proliferative and cytokine cellular responses (p<0.0001) to the EBNA-1 homologous epitope PPPGRRP and the Sm B/B' cross-reactive sequence PPPGMRPP. When immunized with the SLE autoantigen Sm, mCD40-LMP1 Tg mice again exhibited enhanced cellular and humoral immune responses to both Sm and EBNA-1. Cellular immune dysregulation with EBNA-1 immunization in mCD40-LMP1 Tg mice was accompanied by enhanced splenomegaly, increased serum blood urea nitrogen (BUN) and creatinine levels, and elevated anti-dsDNA and antinuclear antibody (ANA) levels (p<0.0001 compared to mCD40 WT mice). However, no evidence of immune-complex glomerulonephritis pathology was noted, suggesting that a combination of EBV and genetic factors may be required to drive lupus-associated renal disease. These data support that the expression of LMP1 in the context of EBNA-1 may interact to increase immune dysregulation that leads to pathogenic, autoantigen-specific lupus inflammation.


Subject(s)
Autoantigens/immunology , Autoimmunity , Epstein-Barr Virus Nuclear Antigens/immunology , Immunity, Cellular , Immunity, Humoral , Lupus Erythematosus, Systemic/immunology , Molecular Mimicry , Viral Matrix Proteins/immunology , snRNP Core Proteins/immunology , Animals , Antibodies, Antinuclear/blood , Autoantigens/administration & dosage , CD40 Antigens/genetics , CD40 Antigens/immunology , CD40 Antigens/metabolism , Cross Reactions , Epitopes , Epstein-Barr Virus Nuclear Antigens/administration & dosage , Immunization , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/genetics , Mice, Inbred C57BL , Mice, Transgenic , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , snRNP Core Proteins/administration & dosage
12.
Front Immunol ; 11: 603029, 2020.
Article in English | MEDLINE | ID: mdl-33537031

ABSTRACT

A synthetic peptide, K-PLP, consisting of 11-unit poly-lysine (K11) linked via polyethylene glycol (PEG) to proteolipid protein epitope (PLP) was synthesized, characterized, and evaluated for efficacy in ameliorating experimental autoimmune encephalomyelitis (EAE) induced by PLP. K-PLP was designed to mimic the cationic nature of the relapsing-remitting multiple sclerosis treatment, glatiramer acetate (GA). With a pI of ~10, GA is able to form visible aggregates at the site of injection via electrostatic interactions with the anionic extracellular matrix. Aggregation further facilitates the retention of GA at the site of injection and draining lymph nodes, which may contribute to its mechanism of action. K-PLP with a pI of ~11, was found to form visible aggregates in the presence of glycosaminoglycans and persist at the injection site and draining lymph nodes in vivo, similar to GA. Additionally, EAE mice treated with K-PLP showed significant inhibition of clinical symptoms compared to free poly-lysine and to PLP, which are the components of K-PLP. The ability of the poly-lysine motif to retain PLP at the injection site, which increased the local exposure of PLP to immune cells may be an important factor affecting drug efficacy.


Subject(s)
Autoantigens/administration & dosage , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Glatiramer Acetate/administration & dosage , Immunosuppressive Agents/administration & dosage , Molecular Mimicry , Peptides/administration & dosage , Animals , Autoantigens/chemistry , Autoantigens/metabolism , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Glatiramer Acetate/metabolism , Immunosuppressive Agents/metabolism , Injections, Subcutaneous , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Peptides/chemical synthesis , Peptides/metabolism , Protein Aggregates , Spleen/drug effects , Spleen/immunology , Spleen/metabolism , Tissue Distribution
13.
Respir Res ; 20(1): 222, 2019 Oct 17.
Article in English | MEDLINE | ID: mdl-31623614

ABSTRACT

Lung cancer (LC) has the highest relative risk of development as a comorbidity of chronic obstructive pulmonary disease (COPD). The molecular mechanisms that mediate chronic inflammation and lung function impairment in COPD have been identified in LC. This suggests the two diseases are more linked than once thought. Emerging data in relation to a key phosphatase, protein phosphatase 2A (PP2A), and its regulatory role in inflammatory and tumour suppression in both disease settings suggests that it may be critical in the progression of COPD to LC. In this review, we uncover the importance of the functional and active PP2A holoenzyme in the context of both diseases. We describe PP2A inactivation via direct and indirect means and explore the actions of two key PP2A endogenous inhibitors, cancerous inhibitor of PP2A (CIP2A) and inhibitor 2 of PP2A (SET), and the role they play in COPD and LC. We explain how dysregulation of PP2A in COPD creates a favourable inflammatory micro-environment and promotes the initiation and progression of tumour pathogenesis. Finally, we highlight PP2A as a druggable target in the treatment of COPD and LC and demonstrate the potential of PP2A re-activation as a strategy to halt COPD disease progression to LC. Although further studies are required to elucidate if PP2A activity in COPD is a causal link for LC progression, studies focused on the potential of PP2A reactivating agents to reduce the risk of LC formation in COPD patients will be pivotal in improving clinical outcomes for both COPD and LC patients in the future.


Subject(s)
Disease Progression , Lung Neoplasms/diagnosis , Lung Neoplasms/enzymology , Protein Phosphatase 2/metabolism , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/enzymology , Animals , Autoantigens/administration & dosage , Humans , Intracellular Signaling Peptides and Proteins/administration & dosage , Lung Neoplasms/drug therapy , Membrane Proteins/administration & dosage , Protein Phosphatase 2/antagonists & inhibitors , Pulmonary Disease, Chronic Obstructive/drug therapy
14.
J Invest Dermatol ; 139(12): 2417-2424.e2, 2019 12.
Article in English | MEDLINE | ID: mdl-31229501

ABSTRACT

Deposition of autoantibodies (α-BP180 and BP230) and complement along the dermal-epidermal-junction is a hallmark of bullous pemphigoid and was shown to be important for pathogenesis. Given the adverse effects of standard treatment (glucocorticoids, immunosuppressants), there is an unmet need for safe and effective therapies. In this phase 1 trial, we evaluated the safety and activity of BIVV009 (sutimlimab, previously TNT009), a targeted C1s inhibitor, in 10 subjects with active or past bullous pemphigoid (NCT02502903). Four weekly 60 mg/kg infusions of BIVV009 proved sufficient for inhibition of the classical complement pathway in all patients, as measured by CH50. C3c deposition along the dermal-epidermal junction was partially or completely abrogated in 4 of 5 patients, where it was present at baseline. BIVV009 was found to be safe and tolerable in this elderly population, with only mild to moderate adverse events reported (e.g., headache, fatigue). One serious adverse event (i.e., fatal cardiac decompensation) occurred at the end of the post-treatment observation period in an 84-year-old patient with a history of diabetes and heart failure, but was deemed unlikely to be related to the study drug. This trial provides the first results with a complement-targeting therapy in bullous pemphigoid, to our knowledge, and supports further studies on BIVV009's efficacy and safety in this population.


Subject(s)
Autoantigens/administration & dosage , Complement C3/metabolism , Complement Pathway, Classical/drug effects , Dermis/pathology , Dystonin/administration & dosage , Epidermis/pathology , Non-Fibrillar Collagens/administration & dosage , Pemphigoid, Bullous/drug therapy , Aged , Aged, 80 and over , Dermis/metabolism , Epidermis/metabolism , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Pemphigoid, Bullous/metabolism , Pemphigoid, Bullous/pathology , Collagen Type XVII
15.
EBioMedicine ; 44: 50-59, 2019 06.
Article in English, French | MEDLINE | ID: mdl-31097410

ABSTRACT

BACKGROUND: Clinical trials on multiple sclerosis with repeated injections of monoclonal antibodies depleting CD4+ T cells have not resulted in much success as a disease therapy. Here, we developed an immunotherapy for EAE in mice by combining a transient depletion of T cells together with the administration of neuron derived peptides. METHODS: EAE was induced in SJL and C57BL/6 mice, by proteolipid protein peptide PLP139-151 (pPLP) and myelin-oligodendrocyte glycoprotein MOG35-55 (pMOG) peptides, respectively. Anti-CD4 and anti-CD8 antibody were injected intraperitoneally before or after peptide immunization. EAE scores were evaluated and histology data from brain and spinal cord were analyzed. Splenocytes were isolated and CD4+, CD4+CD25- and CD4+CD25+ T cells were purified and cultured in the presence of either specific peptides or anti-CD3 antibody and proliferation of T cells as well as cytokines in supernatant were assessed. FINDINGS: This experimental treatment exhibited therapeutic effects on mice with established EAE in pPLP-susceptible SJL mice and pMOG-susceptible C57BL/6 mice. Mechanistically, we revealed that antibody-induced apoptotic T cells triggered macrophages to produce TGFß, and together with administered auto-antigenic peptides, generated antigen-specific Foxp3+ regulatory T cells (Treg cells) in vivo. INTERPRETATION: We successfully developed a specific immunotherapy to EAE by generating autoantigen-specific Treg cells. These findings have overcome the drawbacks of long and repeated depletion of CD4+ T cells, but also obtained long-term immune tolerance, which should have clinical implications for the development of a new effective therapy for multiple sclerosis. FUND: This research was supported by the Intramural Research Program of the NIH, NIDCR.


Subject(s)
Apoptosis/drug effects , Apoptosis/immunology , Autoantigens/administration & dosage , Encephalomyelitis, Autoimmune, Experimental/immunology , Peptides/administration & dosage , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Animals , Autoantigens/immunology , Biomarkers/metabolism , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Immune Tolerance , Immunologic Factors , Immunotherapy , Lymphocyte Activation , Mice , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Peptides/immunology , Phagocytes/drug effects , Phagocytes/immunology , Phagocytes/metabolism , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Treatment Outcome
16.
J Pharmacol Sci ; 139(3): 193-200, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30827890

ABSTRACT

Ischemia/reperfusion (I/R)-induced oxidative stress is a serious clinical problem in the reperfusion therapy for ischemic diseases. Tumstatin is an endogenous bioactive peptide cleaved from type IV collagen α3 chain. We previously reported that T3 peptide, an active subfragment of tumstatin, exerts cytoprotective effects on H2O2-induced apoptosis through the inhibition of intracellular reactive oxygen species (ROS) production in H9c2 cardiomyoblasts. In this study, we investigated whether T3 peptide has cardioprotective effects against I/R injury by using in vitro and ex vivo experimental models. H9c2 cardiomyoblasts were stimulated with oxygen and glucose deprivation (OGD) for 12 h followed by reoxygenation for 1-8 h (OGD/R; in vitro model). The cells were treated with T3 peptide (30-1000 ng/ml) during OGD. Ten minutes after the pre-perfusion of T3 peptide (300 ng/ml), Langendorff perfused rat hearts were exposed to ischemia for 30 min followed by reperfusion for 1 h (ex vivo model). T3 peptide inhibited OGD/R-induced apoptosis through the inhibition of mitochondrial ROS production and dysfunction in H9c2 cardiomyoblasts. T3 peptide also prevented I/R-induced cardiac dysfunction, arrhythmia and myocardial infarction in the perfused rat heart. In conclusion, we for the first time demonstrated that T3 peptide exerts cardioprotective effects against I/R injury.


Subject(s)
Apoptosis/drug effects , Autoantigens/administration & dosage , Cardiotonic Agents/administration & dosage , Collagen Type IV/administration & dosage , Myocardial Reperfusion Injury/drug therapy , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/prevention & control , Autoantigens/chemistry , Autoantigens/pharmacology , Cardiotonic Agents/pharmacology , Cell Line , Collagen Type IV/chemistry , Collagen Type IV/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Myocardial Infarction/etiology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/complications , Peptides/administration & dosage , Peptides/pharmacology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
17.
Int J Pharm ; 562: 303-312, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30910633

ABSTRACT

Antigen specific immunotherapy aims to tolerise patients to specific autoantigens that are responsible for the pathology of an autoimmune disease. Immune tolerance is generated in conditions where the immune response is suppressed and thus gold nanoparticles (AuNPs) are an attractive drug delivery platform due to their anti-inflammatory effects and their potential to facilitate temporal and spatial delivery of a peptide autoantigen in conjunction with pro-tolerogenic elements. In this study we have covalently attached an autoantigen, currently under clinical evaluation for the treatment of type 1 diabetes (PIC19-A3 peptide), to AuNPs to create nanoscale (<5 nm), negatively charged (-40 to -60 mV) AuNP-peptide complexes for immunotherapy. We also employ a clinically approved microneedle delivery system, MicronJet600, to facilitate minimally-invasive intradermal delivery of the nanoparticle constructs to target skin-resident antigen presenting cells, which are known to be apposite target cells for immunotherapy. The AuNP-peptide complexes remain physically stable upon extrusion through microneedles and when delivered into ex vivo human skin they are able to diffuse rapidly and widely throughout the dermis (their site of deposition) and, perhaps more surprisingly, the overlying epidermal layer. Intracellular uptake was extensive, with Langerhans cells proving to be the most efficient cells at internalising the AuNP-peptide complex (94% of the local population within the treated region of skin). In vitro studies showed that uptake of the AuNP-peptide complexes by dendritic cells reduced the capacity of these cells to activate naïve T cells. This indicator of biological functionality encourages further development of the AuNP-peptide formulation, which is now being evaluated in clinical trials.


Subject(s)
Autoantigens/administration & dosage , Gold/administration & dosage , Immunotherapy , Metal Nanoparticles/administration & dosage , Peptides/administration & dosage , Skin/metabolism , Adult , Aged , Aged, 80 and over , Biological Transport , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Humans , Injections, Intradermal , Middle Aged , Skin/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
18.
PLoS One ; 14(3): e0214379, 2019.
Article in English | MEDLINE | ID: mdl-30908554

ABSTRACT

Autoantigen-specific methods to prevent and treat Type 1 Diabetes (T1D) carry high hopes to permanently cure this disease, but have largely failed in clinical trials. One suggested approach to increase the efficacy of islet antigen-specific vaccination is to combine it with a modulator of the T cell response, with the goal of reducing effector differentiation and promoting regulatory T cells (Tregs). Here we asked if addition of antibodies that block the IL-7/IL-7Rα pathway altered the T cell response to islet antigen vaccination and prevented T1D in non-obese diabetic (NOD) mice. Anti-IL-7Rα monoclonal antibodies (mAbs) reduced the numbers of islet antigen-specific T cells generated after vaccination with islet peptides and alum. However, addition of anti-IL-7Rα antibodies to peptide/alum vaccination unexpectedly increased non-specific IFN-γ, IL-2 and IL-10 cytokine production and did not result in improved prevention of T1D onset. In a second approach, we used a conjugate vaccine to deliver islet autoantigens, using Keyhole Limpet Hemocyanin (KLH) as a carrier. Islet antigen-KLH vaccination led to a significant expansion of antigen-specific Tregs and delayed diabetes onset in NOD mice. These outcomes were not further improved by addition of anti-IL-7Rα antibodies. To the contrary, blocking IL-7Rα during vaccination led to non-specific cytokine production and reduced the efficacy of a KLH-conjugated vaccine to prevent T1D. Our study thus revealed that adding anti-IL-7Rα antibodies during autoantigen immunization did not improve the efficacy of such vaccinations to prevent T1D, despite altering some aspects of the T cell response in a potentially advantageous way. Further refinement of this approach will be required to separate the beneficial from the adverse effects of anti-IL-7Rα antibodies to treat autoimmune disease.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Autoantigens/administration & dosage , Cytokines/metabolism , Diabetes Mellitus, Type 1/immunology , Interleukin-7 Receptor alpha Subunit/immunology , Animals , Antibodies, Monoclonal/pharmacology , Autoantigens/immunology , Cell Proliferation , Diabetes Mellitus, Type 1/drug therapy , Disease Models, Animal , Female , Immunization , Immunotherapy , Mice , Mice, Inbred NOD , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Vaccination
19.
Mol Pharm ; 16(2): 607-617, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30615457

ABSTRACT

Contemporary approaches to treating autoimmune diseases like multiple sclerosis broadly modulate the immune system and leave patients susceptible to severe adverse effects. Antigen-specific immunotherapies (ASIT) offer a unique opportunity to selectively suppress autoreactive cell populations but have suffered from marginal efficacy even when employing traditional adjuvants to improve delivery. The development of immunologically active antigen delivery vehicles could potentially increase the clinical success of antigen-specific immunotherapies. An emulsion of the antioxidant tocopherol delivering an epitope of proteolipid protein autoantigen (PLP139-151) yielded significant efficacy in mice with experimental autoimmune encephalomyelitis (EAE). In vitro studies indicated tocopherol emulsions reduced oxidative stress in antigen-presenting cells. Ex vivo analysis revealed that tocopherol emulsions shifted cytokine responses in EAE splenocytes. In addition, IgG responses against PLP139-151 were increased in mice treated with tocopherol emulsions delivering the antigen, suggesting a possible skew in immunity. Overall, tocopherol emulsions provide a functional delivery vehicle for ASIT capable of ameliorating autoimmunity in a murine model.


Subject(s)
Autoantigens/therapeutic use , Emulsions/chemistry , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Tocopherols/chemistry , Tocopherols/therapeutic use , Animals , Autoantigens/administration & dosage , Cytokines/metabolism , Female , Immune Tolerance/drug effects , Immunotherapy/methods , Mice , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/pathogenicity , Spleen/cytology
20.
Front Immunol ; 9: 2871, 2018.
Article in English | MEDLINE | ID: mdl-30574145

ABSTRACT

Breaking tolerance is a key event leading to autoimmunity, but the exact mechanisms responsible for this remain uncertain. Here we show that the alarmin IL-33 is able to drive the generation of autoantibodies through induction of the B cell survival factor BAFF. A temporary, short-term increase in IL-33 results in a primary (IgM) response to self-antigens. This transient DNA-specific autoantibody response was dependent on the induction of BAFF. Notably, radiation resistant cells and not myeloid cells, such as neutrophils or dendritic cells were the major source of BAFF and were critical in driving the autoantibody response. Chronic exposure to IL-33 elicited dramatic increases in BAFF levels and resulted in elevated numbers of B and T follicular helper cells as well as germinal center formation. We also observed class-switching from an IgM to an IgG DNA-specific autoantibody response. Collectively, the results provide novel insights into a potential mechanism for breaking immune-tolerance via IL-33-mediated induction of BAFF.


Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , Autoimmune Diseases/immunology , B-Cell Activating Factor/metabolism , Immune Tolerance , Interleukin-33/immunology , Animals , Autoantigens/administration & dosage , B-Cell Activating Factor/genetics , B-Cell Activating Factor/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cells, Cultured , Dendritic Cells , Disease Models, Animal , Humans , Immunoglobulin M/immunology , Interleukin-33/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils , Primary Cell Culture , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...