Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Nat Immunol ; 25(6): 1083-1096, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38816616

ABSTRACT

Current prophylactic human immunodeficiency virus 1 (HIV-1) vaccine research aims to elicit broadly neutralizing antibodies (bnAbs). Membrane-proximal external region (MPER)-targeting bnAbs, such as 10E8, provide exceptionally broad neutralization, but some are autoreactive. Here, we generated humanized B cell antigen receptor knock-in mouse models to test whether a series of germline-targeting immunogens could drive MPER-specific precursors toward bnAbs. We found that recruitment of 10E8 precursors to germinal centers (GCs) required a minimum affinity for germline-targeting immunogens, but the GC residency of MPER precursors was brief due to displacement by higher-affinity endogenous B cell competitors. Higher-affinity germline-targeting immunogens extended the GC residency of MPER precursors, but robust long-term GC residency and maturation were only observed for MPER-HuGL18, an MPER precursor clonotype able to close the affinity gap with endogenous B cell competitors in the GC. Thus, germline-targeting immunogens could induce MPER-targeting antibodies, and B cell residency in the GC may be regulated by a precursor-competitor affinity gap.


Subject(s)
Antibody Affinity , B-Lymphocytes , Germinal Center , HIV Antibodies , HIV-1 , Germinal Center/immunology , Animals , Mice , Humans , B-Lymphocytes/immunology , HIV-1/immunology , HIV Antibodies/immunology , Antibody Affinity/immunology , Antibodies, Neutralizing/immunology , HIV Infections/immunology , AIDS Vaccines/immunology , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, B-Cell/immunology , Gene Knock-In Techniques , Mice, Transgenic , Broadly Neutralizing Antibodies/immunology , Mice, Inbred C57BL
2.
Nat Immunol ; 25(6): 1073-1082, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38816615

ABSTRACT

A key barrier to the development of vaccines that induce broadly neutralizing antibodies (bnAbs) against human immunodeficiency virus (HIV) and other viruses of high antigenic diversity is the design of priming immunogens that induce rare bnAb-precursor B cells. The high neutralization breadth of the HIV bnAb 10E8 makes elicitation of 10E8-class bnAbs desirable; however, the recessed epitope within gp41 makes envelope trimers poor priming immunogens and requires that 10E8-class bnAbs possess a long heavy chain complementarity determining region 3 (HCDR3) with a specific binding motif. We developed germline-targeting epitope scaffolds with affinity for 10E8-class precursors and engineered nanoparticles for multivalent display. Scaffolds exhibited epitope structural mimicry and bound bnAb-precursor human naive B cells in ex vivo screens, protein nanoparticles induced bnAb-precursor responses in stringent mouse models and rhesus macaques, and mRNA-encoded nanoparticles triggered similar responses in mice. Thus, germline-targeting epitope scaffold nanoparticles can elicit rare bnAb-precursor B cells with predefined binding specificities and HCDR3 features.


Subject(s)
AIDS Vaccines , Antibodies, Neutralizing , HIV Antibodies , HIV Envelope Protein gp41 , HIV Infections , HIV-1 , Macaca mulatta , Animals , Humans , HIV Envelope Protein gp41/immunology , HIV Antibodies/immunology , Mice , AIDS Vaccines/immunology , Antibodies, Neutralizing/immunology , HIV-1/immunology , HIV Infections/immunology , HIV Infections/prevention & control , HIV Infections/virology , Vaccination , Broadly Neutralizing Antibodies/immunology , B-Lymphocytes/immunology , Nanoparticles/chemistry , Female , Complementarity Determining Regions/immunology , Epitopes/immunology
3.
Immunity ; 57(5): 1141-1159.e11, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38670113

ABSTRACT

Broadly neutralizing antibodies (bnAbs) targeting the hemagglutinin (HA) stem of influenza A viruses (IAVs) tend to be effective against either group 1 or group 2 viral diversity. In rarer cases, intergroup protective bnAbs can be generated by human antibody paratopes that accommodate the conserved glycan differences between the group 1 and group 2 stems. We applied germline-engaging nanoparticle immunogens to elicit a class of cross-group bnAbs from physiological precursor frequency within a humanized mouse model. Cross-group protection depended on the presence of the human bnAb precursors within the B cell repertoire, and the vaccine-expanded antibodies enriched for an N55T substitution in the CDRH2 loop, a hallmark of the bnAb class. Structurally, this single mutation introduced a flexible fulcrum to accommodate glycosylation differences and could alone enable cross-group protection. Thus, broad IAV immunity can be expanded from the germline repertoire via minimal antigenic input and an exceptionally simple antibody development pathway.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Influenza A virus , Influenza Vaccines , Orthomyxoviridae Infections , Vaccination , Animals , Mice , Humans , Antibodies, Viral/immunology , Influenza Vaccines/immunology , Influenza A virus/immunology , Antibodies, Neutralizing/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Amino Acid Substitution , B-Lymphocytes/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Broadly Neutralizing Antibodies/immunology
5.
EMBO J ; 40(2): e105926, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33258500

ABSTRACT

B-cell receptor (BCR) knock-in (KI) mouse models play an important role in vaccine development and fundamental immunological studies. However, the time required to generate them poses a bottleneck. Here we report a one-step CRISPR/Cas9 KI methodology to combine the insertion of human germline immunoglobulin heavy and light chains at their endogenous loci in mice. We validate this technology with the rapid generation of three BCR KI lines expressing native human precursors, instead of computationally inferred germline sequences, to HIV broadly neutralizing antibodies. We demonstrate that B cells from these mice are fully functional: upon transfer to congenic, wild type mice at controlled frequencies, such B cells can be primed by eOD-GT8 60mer, a germline-targeting immunogen currently in clinical trials, recruited to germinal centers, secrete class-switched antibodies, undergo somatic hypermutation, and differentiate into memory B cells. KI mice expressing functional human BCRs promise to accelerate the development of vaccines for HIV and other infectious diseases.


Subject(s)
B-Lymphocytes/metabolism , CRISPR-Cas Systems/genetics , Receptors, Antigen, B-Cell/metabolism , Animals , B-Lymphocytes/immunology , Broadly Neutralizing Antibodies/immunology , CRISPR-Cas Systems/immunology , Cell Line , Gene Knock-In Techniques/methods , Germinal Center/immunology , Germinal Center/metabolism , HEK293 Cells , HIV-1/immunology , Humans , Male , Mice , Mice, Inbred C57BL , Models, Animal , Receptors, Antigen, B-Cell/immunology
6.
Crit Rev Biochem Mol Biol ; 55(6): 555-575, 2020 12.
Article in English | MEDLINE | ID: mdl-32933340

ABSTRACT

Receptor for advanced glycation end products (RAGE) is an immunoglobulin-like receptor present on cell surface. RAGE binds to an array of structurally diverse ligands, acts as a pattern recognition receptor (PRR) and is expressed on cells of different origin performing different functions. RAGE ligation leads to the initiation of a cascade of signaling events and is implicated in diseases, such as inflammation, cancer, diabetes, vascular dysfunctions, retinopathy, and neurodegenerative diseases. Because of the significant involvement of RAGE in the progression of numerous diseases, RAGE signaling has been targeted through use of inhibitors and anti-RAGE antibodies as a treatment strategy and therapy. Here in this review, we have summarized the physical and physiological aspects of RAGE biology in mammalian system and the importance of targeting this molecule in the treatment of various RAGE mediated pathologies. Highlights Receptor for advanced glycation end products (RAGE) is a member of immunoglobulin superfamily of receptors and involved in many pathophysiological conditions. RAGE ligation with its ligands leads to initiation of distinct signaling cascades and activation of numerous transcription factors. Targeting RAGE signaling through inhibitors and anti-RAGE antibodies can be promising treatment strategy.


Subject(s)
Diabetes Mellitus/metabolism , Glycation End Products, Advanced/metabolism , Receptor for Advanced Glycation End Products/metabolism , Animals , Diabetes Mellitus/genetics , Glycation End Products, Advanced/genetics , Humans , Receptor for Advanced Glycation End Products/genetics , Signal Transduction/genetics , Signal Transduction/physiology
7.
Cell Commun Signal ; 18(1): 170, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33109194

ABSTRACT

BACKGROUND: Receptor for advanced glycation end products (RAGE) is a multi-ligand transmembrane receptor of the immunoglobulin superfamily. Lysophosphatidic acid (LPA) is a ligand for RAGE and is involved in physiological and pathophysiological conditions including cancer. However, RAGE-LPA axis is unexplored in lung and mammary cancer. METHODS: RAGE was silenced in A549, MDA MB-231 and MCF7 using RAGE shRNA. For in vitro tumorigenesis, we performed wound healing, colony formation, cell proliferation and invasion assays. Evaluation of expression of oncogenes, EMT markers and downstream signaling molecules was done by using western blot and immunohistochemistry. For subcellular expression of RAGE, immunofluorescence was done. In vivo tumorigenesis was assessed by intraperitoneal injection of cancer cells in nude mice. RESULTS: Here we show RAGE mediated profound increase in proliferation, migration and invasion of lung and mammary cancer cells via LPA in Protein kinase B (PKB) dependent manner. LPA mediated EMT transition is regulated by RAGE. In vivo xenograft results show significance of RAGE in LPA mediated lung and mammary tumor progression, angiogenesis and immune cell infiltration to tumor microenvironment. CONCLUSION: Our results establish the significance and involvement of RAGE in LPA mediated lung and mammary tumor progression and EMT transition via RAGE. RAGE-LPA axis may be a therapeutic target in lung and mammary cancer treatment strategies. Video Abstract.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Lung Neoplasms/pathology , Lysophospholipids/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction , Tumor Microenvironment , Animals , Breast Neoplasms/immunology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Gene Knockdown Techniques , Humans , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Xenograft Model Antitumor Assays
8.
Blood ; 129(9): 1177-1183, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28069607

ABSTRACT

Monocytes and macrophages represent critical arms of the innate immune system and are considered regulators and effectors of inflammation and the innate immune response. Monocytes can mobilize from bone marrow, traffic to their required destination, and differentiate into effector cells, depending on the local tissue environment, to perform multiple roles during infection or inflammation, making them important components of body's immune defense. Macrophages have diverse roles in tissue homeostasis, development, and tissue repair following injury. Adult bone marrow monocytes can give rise to tissue-resident macrophages during infection or inflammatory reactions, besides self-replication of tissue resident macrophages. Lysophosphatidic acid (LPA), a lipid by-product of autotaxin activity, is involved in cancer, vascular defects, and neural tissue, but is largely unexplored in immune system. Here, we reveal an unexpected function of LPA that transfigures CD11b+ murine monocytes into F4/80+ macrophages. LPA-stimulated Akt/mTOR signaling is critical for LPA-mediated macrophage development in mice. Additionally, transcriptome analysis reveals that PPARγ is the key transcriptional regulator in the development of LPA-induced macrophages. In humans, LPA mediates macrophage formation following similar pathways. These findings identify a critical role for LPA in regulating innate immune system.


Subject(s)
Cell Differentiation/immunology , Lysophospholipids/pharmacology , Monocytes/cytology , Animals , Cell Differentiation/drug effects , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Immunity, Innate/drug effects , Immunity, Innate/immunology , Immunoblotting , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/immunology , PPAR gamma/immunology
9.
Vasc Med ; 20(3): 212-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25808570

ABSTRACT

We investigated treatment with a receptor for advanced glycation endproduct (RAGE) blocking antibody on angiogenic response to hind limb ischemia in diabetic mice. Streptozotocin treated C57BL/6 mice received either murine monoclonal anti-RAGE F(ab')2 intraperitoneally (n=10) or saline (n=9) for 9 weeks. Diabetic plus 10 non-diabetic C57BL/6 mice underwent left femoral artery ligation and 5 days later angiogenesis imaging with (99m)Tc-Arg-Gly-Asp (RGD) nanoSPECT/CT. Twenty-four days later, hind limb blood flow was measured with ultrasound, the mice were euthanized, and tissue was taken for immunohistochemistry. The angiogenic imaging signal in ischemic limbs was higher in RAGE-ab treated versus saline treated mice at day 5 (3.1±1.4 vs 1.68±0.35, p=0.02) and blood flow was higher at day 24 (1.49±0.5 vs 0.61±0.39, p=0.04). Immunohistochemistry of ischemic muscles showed greater capillary density in the RAGE-ab treated group versus the vehicle-treated group (p<0.001) (NS from non-diabetic mice). In conclusion, treatment with anti-RAGE F(ab')2 in diabetic mice improves neovascularization in the ischemic leg.


Subject(s)
Antibodies/therapeutic use , Hindlimb/blood supply , Ischemia/drug therapy , Neovascularization, Physiologic/drug effects , Receptor for Advanced Glycation End Products/immunology , Animals , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1/complications , Diabetic Angiopathies/drug therapy , Diabetic Angiopathies/etiology , Femoral Artery , Ligation , Male , Mice , Mice, Inbred C57BL
10.
Biochemistry ; 53(20): 3327-35, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24824951

ABSTRACT

Diabetes-induced hyperglycemia increases the extracellular concentration of methylglyoxal. Methylglyoxal-derived hydroimidazolones (MG-H) form advanced glycation end products (AGEs) that accumulate in the serum of diabetic patients. The binding of hydroimidozolones to the receptor for AGEs (RAGE) results in long-term complications of diabetes typified by vascular and neuronal injury. Here we show that binding of methylglyoxal-modified albumin to RAGE results in signal transduction. Chemically synthesized peptides containing hydroimidozolones bind specifically to the V domain of RAGE with nanomolar affinity. The solution structure of an MG-H1-V domain complex revealed that the hydroimidazolone moiety forms multiple contacts with a positively charged surface on the V domain. The high affinity and specificity of hydroimidozolones binding to the V domain of RAGE suggest that they are the primary AGE structures that give rise to AGEs-RAGE pathologies.


Subject(s)
Glycation End Products, Advanced/chemistry , Glycation End Products, Advanced/metabolism , Pyruvaldehyde/chemistry , Pyruvaldehyde/metabolism , Receptors, Immunologic/chemistry , Receptors, Immunologic/metabolism , Cell Line , Humans , Protein Structure, Secondary , Protein Structure, Tertiary , Receptor for Advanced Glycation End Products , Signal Transduction/physiology
11.
Sci Immunol ; 9(95): eadn0622, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38753808

ABSTRACT

Germline-targeting (GT) protein immunogens to induce VRC01-class broadly neutralizing antibodies (bnAbs) to the CD4-binding site of the HIV envelope (Env) have shown promise in clinical trials. Here, we preclinically validated a lipid nanoparticle-encapsulated nucleoside mRNA (mRNA-LNP) encoding eOD-GT8 60mer as a soluble self-assembling nanoparticle in mouse models. In a model with three humanized B cell lineages bearing distinct VRC01-precursor B cell receptors (BCRs) with similar affinities for eOD-GT8, all lineages could be simultaneously primed and undergo diversification and affinity maturation without exclusionary competition. Boosts drove precursor B cell participation in germinal centers; the accumulation of somatic hypermutations, including in key VRC01-class positions; and affinity maturation to boost and native-like antigens in two of the three precursor lineages. We have preclinically validated a prime-boost regimen of soluble self-assembling nanoparticles encoded by mRNA-LNP, demonstrating that multiple lineages can be primed, boosted, and diversified along the bnAb pathway.


Subject(s)
Broadly Neutralizing Antibodies , Nanoparticles , RNA, Messenger , Animals , Mice , Humans , RNA, Messenger/immunology , RNA, Messenger/genetics , Nanoparticles/chemistry , Broadly Neutralizing Antibodies/immunology , HIV Antibodies/immunology , Lipids/immunology , HIV Infections/immunology , AIDS Vaccines/immunology , Antibodies, Neutralizing/immunology , HIV-1/immunology , Female , Antibodies, Monoclonal , Liposomes
12.
Science ; 384(6697): eadk0582, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38753770

ABSTRACT

Germline-targeting (GT) HIV vaccine strategies are predicated on deriving broadly neutralizing antibodies (bnAbs) through multiple boost immunogens. However, as the recruitment of memory B cells (MBCs) to germinal centers (GCs) is inefficient and may be derailed by serum antibody-induced epitope masking, driving further B cell receptor (BCR) modification in GC-experienced B cells after boosting poses a challenge. Using humanized immunoglobulin knockin mice, we found that GT protein trimer immunogen N332-GT5 could prime inferred-germline precursors to the V3-glycan-targeted bnAb BG18 and that B cells primed by N332-GT5 were effectively boosted by either of two novel protein immunogens designed to have minimum cross-reactivity with the off-target V1-binding responses. The delivery of the prime and boost immunogens as messenger RNA lipid nanoparticles (mRNA-LNPs) generated long-lasting GCs, somatic hypermutation, and affinity maturation and may be an effective tool in HIV vaccine development.


Subject(s)
AIDS Vaccines , Broadly Neutralizing Antibodies , Germinal Center , HIV Antibodies , HIV-1 , Immunization, Secondary , Nanoparticles , mRNA Vaccines , Animals , Humans , Mice , AIDS Vaccines/immunology , B-Lymphocytes/immunology , Broadly Neutralizing Antibodies/immunology , Cross Reactions , Gene Knock-In Techniques , Germinal Center/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/genetics , HIV Infections/immunology , HIV Infections/prevention & control , HIV-1/immunology , HIV-1/genetics , Liposomes , Memory B Cells/immunology , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/genetics , Somatic Hypermutation, Immunoglobulin , mRNA Vaccines/immunology , Female , Mice, Inbred C57BL
13.
STAR Protoc ; 3(1): 101111, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35118424

ABSTRACT

The skeletal muscle system is the major organ associated with movement of the body. Myogenesis and regeneration induced post-injury contribute to muscle formation and maintenance. Here, we provide detailed protocol for the accelerated repair of injured skeletal muscles and generation of hypertrophic muscle fibers. This protocol includes cardiotoxin induced muscle injury and also describes isolation of satellite cells from skeletal muscle tissues of mice. This protocol can be used to study the mechanisms associated with accelerated muscle repair and hypertrophy. For complete details on the use and execution of this protocol, please refer to Ray et al. (2021).


Subject(s)
Hypertrophy/physiopathology , Muscle, Skeletal/physiology , Muscular Diseases/physiopathology , Regeneration , Animals , Mice , Mice, Inbred C57BL
14.
Cell Rep ; 34(9): 108809, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33657371

ABSTRACT

Muscle differentiation is a multifaceted and tightly controlled process required for the formation of skeletal muscle fibers. Satellite cells are the direct cellular contributors to muscle repair in injuries or disorders. Here, we show that autotaxin (Atx) expression and activity is required for satellite cell differentiation. Conditional ablation of Atx or its pharmacological inhibition impairs muscle repair. Mechanistically, we identify LPAR1 as the key receptor in Atx-LPA signaling. Myogenic gene array and pathway analysis identified that Atx-LPA signaling activates ribosomal protein S6 kinase (S6K), an mTOR-dependent master regulator of muscle cell growth via LPAR1. Furthermore, Atx transgenic mice show muscle hypertrophic effects and accelerated regeneration. Intramuscular injections of Atx/LPA show muscle hypertrophy. In addition, the regulatory effects of Atx on differentiation are conserved in human myoblasts. This study identifies Atx as a critical master regulator in murine and human muscles, identifying a promising extracellular ligand in muscle formation, regeneration, and hypertrophy.


Subject(s)
Lysophospholipids/metabolism , Muscle Development , Muscle, Skeletal/metabolism , Phosphoric Diester Hydrolases/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Regeneration , Satellite Cells, Skeletal Muscle/metabolism , Animals , Cell Line , Female , Gene Expression Regulation , Humans , Hypertrophy , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Phosphoric Diester Hydrolases/genetics , Receptors, Lysophosphatidic Acid/genetics , Ribosomal Protein S6 Kinases/metabolism , Satellite Cells, Skeletal Muscle/pathology , Signal Transduction , Skeletal Muscle Enlargement , TOR Serine-Threonine Kinases/metabolism
15.
Cell Death Dis ; 12(11): 1012, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34711805

ABSTRACT

Melanoma originates from melanin-producing cells called melanocytes. Melanoma poses a great risk because of its rapid ability to spread and invade new organs. Cellular metastasis involves alteration in the gene expression profile and their transformation from epithelial to mesenchymal state. Despite of several advances, metastatic melanoma being a key cause of therapy failure and mortality remains poorly understood. p32 has been found to be involved in various physiological and pathophysiological conditions. However, the role of p32 in melanoma progression and metastasis remains underexplored. Here, we identify the role of p32 in the malignancy of both murine and human melanoma. p32 knockdown leads to reduced cell proliferation, migration, and invasion in murine and human melanoma cells. Furthermore, p32 promotes in vitro tumorigenesis, inducing oncogenes and EMT markers. Mechanistically, we show p32 regulates tumorigenic and metastatic properties through the Akt/PKB signaling pathway in both murine and human melanoma. Furthermore, p32 silencing attenuates melanoma tumor progression and lung metastasis in vivo, modulating the tumor microenvironment by inhibiting the angiogenesis, infiltration of macrophages, and leukocytes in mice. Taken together, our findings identify that p32 drives melanoma progression, metastasis, and regulates the tumor microenvironment. p32 can be a target of a novel therapeutic approach in the regulation of melanoma progression and metastasis.


Subject(s)
Carrier Proteins/adverse effects , Epithelial-Mesenchymal Transition/genetics , Melanoma/genetics , Mitochondrial Proteins/adverse effects , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Movement , Cell Proliferation , Disease Progression , Humans , Melanoma/mortality , Melanoma/physiopathology , Mice , Neoplasm Metastasis , Signal Transduction , Survival Analysis , Transfection , Tumor Microenvironment
16.
Med Oncol ; 37(10): 88, 2020 Sep 09.
Article in English | MEDLINE | ID: mdl-32902730

ABSTRACT

Non-muscle myosin IIA heavy chain (MYH9) has been implicated in many physiological and pathological functions including cell adhesion, polarity, motility to cancer. However, its role in melanoma remains unexplored. The aim of our study was to evaluate the role of MYH9 in melanoma tumor development and metastasis and further to find out the potential underlying mechanisms. In this study, we evaluated the in vitro migratory and invasive properties and in vivo tumor development and metastasis in C57BL/6 mice by silencing MYH9 in B16F10 melanoma cells. Knocking down MYH9 enhanced migration and invasiveness of B16F10 cells in vitro. Furthermore, MYH9 silencing accelerated tumor growth and metastasis in melanoma subcutaneous and intravenous mouse models. Next, oncogenes analysis revealed epithelial-mesenchymal transition and Erk signaling pathway are being regulated with MYH9 expression. Finally, MYH9 silencing in B16F10 cells modulates the tumor microenvironment by manipulating the leukocytes and macrophages infiltration in tumors. These findings established the opposing role of MYH9 as a tumor suppressor in melanoma suggesting specific MYH9 based approaches in therapeutics.


Subject(s)
Melanoma, Experimental/pathology , Myosin Heavy Chains/metabolism , Tumor Microenvironment/physiology , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation/physiology , Epithelial-Mesenchymal Transition/physiology , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness/pathology
17.
Biochimie ; 154: 55-61, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30076903

ABSTRACT

Receptor for Advanced Glycation End product (RAGE) is a multiligand receptor implicated in diverse pathological conditions such as diabetes, atherosclerosis, cancer and neural diseases. Extracellular, RAGE consists of V, C1 and C2 domains. Here, we show RAGE exists as a monomer in equilibrium with a fraction of a covalently linked dimer of monomers via its V domain through cysteine. In order to understand the functional implication of this dimer, we examined the binding capacity and functional potential of RAGE dimer via advanced glycation end products (AGEs) which shows enhanced binding capacity towards V domain, ERK phosphorylation, cytokine release and actin polymerization ability of the dimeric form for AGEs compared with the reduced monomeric form. Our data, suggests that the dimeric state of RAGE controls its function and ligand mediated signaling which may play important role in RAGE mediated various diseases.


Subject(s)
Cysteine/metabolism , Disulfides/metabolism , Protein Multimerization , Receptor for Advanced Glycation End Products/metabolism , A549 Cells , Animals , Cysteine/chemistry , Cysteine/genetics , Disulfides/chemistry , Glycation End Products, Advanced/metabolism , Humans , Mice , Protein Domains , Receptor for Advanced Glycation End Products/chemistry , Receptor for Advanced Glycation End Products/genetics
18.
Neurosci Biobehav Rev ; 62: 48-55, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26724598

ABSTRACT

RAGE, the receptor of advanced glycation end-products, is thought to be one of the potential contributors to the neurodegeneration. It has been shown that RAGE activation triggers an increase in proinflammatory molecules, oxidative stressors and cytokines. RAGE involvement has been documented in the pathogenesis of a number of neurodegenerative diseases such amyotrophic lateral sclerosis (ALS), Alzheimer's, Parkinson's, Huntington's, Creutzfeld-Jakob' diseases and various neurodegenerative conditions such as diabetic neuropathy, familial amyloid polyneuropathy, Charcot neuroarthropathy and vasculitic neuropathy. Although the detailed mechanisms of RAGE contribution to the neurodegeneration remains unclear, studies indicate that RAGE detrimental actions are exerted via its binding to the pro-inflammatory ligands such as advanced glycation end-products, S100/calgranulin and amphoterin and subsequent activation of downstream regulatory pathways such as NF-κB, STAT and JKN pathways. Here, in this review we attempt to shed light onto molecular events and pathological pathways involved in neuroinflammation, neurodegeneration and its emerging role in the pathogenesis of amyotrophic lateral sclerosis (ALS)--a progressive and fatal neurodegenerative disorder, summarizing current knowledge and the prospect of RAGE in the pathogenesis of this disastrous disease.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Inflammation/metabolism , Neurodegenerative Diseases/physiopathology , Signal Transduction/physiology , Animals , Carrier Proteins/metabolism , Humans , Neurodegenerative Diseases/immunology , Oxidative Stress/immunology , Oxidative Stress/physiology
19.
Rev Neurosci ; 26(6): 691-8, 2015.
Article in English | MEDLINE | ID: mdl-26226128

ABSTRACT

This review, for the first time, aims to summarize the current knowledge in the emerging field of RAGE (receptor for advanced glycation end-products) studies in neurodegeneration and neurodegenerative diseases. RAGE, a member of the multiligand cell surface immunoglobulin family, has been implicated in numerous pathological conditions - from diabetes and cardiovascular diseases to tumors and neurodegenerative disorders, such as Alzheimer's disease, familial amyloid polyneuropathy, diabetic neuropathy, Parkinson's disease, and Huntington's disease. Until now, the detailed mechanisms of the contribution of RAGE to neurodegeneration remain elusive; however, mounting evidence suggests that its detrimental actions are triggered by its ligand interactions and contribute to increased neuroinflammation, neuronal degeneration, and apoptosis. Deciphering the role of RAGE in neurodegenerative disorders will be a milestone in our basic understanding of the mechanisms involved in the pathogenesis of neurodegeneration, helping to delineate molecular links between complex RAGE signaling pathways and neuronal dysfunction and neurodegeneration.


Subject(s)
Glycation End Products, Advanced/metabolism , Neurodegenerative Diseases/metabolism , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction/physiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL