Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.452
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 24(4): 664-675, 2023 04.
Article in English | MEDLINE | ID: mdl-36849745

ABSTRACT

Antigen-specific CD8+ T cell accumulation in tumors is a prerequisite for effective immunotherapy, and yet the mechanisms of lymphocyte transit are not well defined. Here we show that tumor-associated lymphatic vessels control T cell exit from tumors via the chemokine CXCL12, and intratumoral antigen encounter tunes CXCR4 expression by effector CD8+ T cells. Only high-affinity antigen downregulates CXCR4 and upregulates the CXCL12 decoy receptor, ACKR3, thereby reducing CXCL12 sensitivity and promoting T cell retention. A diverse repertoire of functional tumor-specific CD8+ T cells, therefore, exit the tumor, which limits the pool of CD8+ T cells available to exert tumor control. CXCR4 inhibition or loss of lymphatic-specific CXCL12 boosts T cell retention and enhances tumor control. These data indicate that strategies to limit T cell egress might be an approach to boost the quantity and quality of intratumoral T cells and thereby response to immunotherapy.


Subject(s)
Lymphatic Vessels , Neoplasms , Humans , CD8-Positive T-Lymphocytes , Receptors, CXCR4/metabolism , Neoplasms/therapy , Neoplasms/pathology , Lymphatic Vessels/metabolism , Immunotherapy
2.
Cell ; 183(5): 1282-1297.e18, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33098771

ABSTRACT

Classically considered short-lived and purely defensive leukocytes, neutrophils are unique in their fast and moldable response to stimulation. This plastic behavior may underlie variable and even antagonistic functions during inflammation or cancer, yet the full spectrum of neutrophil properties as they enter healthy tissues remains unexplored. Using a new model to track neutrophil fates, we found short but variable lifetimes across multiple tissues. Through analysis of the receptor, transcriptional, and chromatin accessibility landscapes, we identify varying neutrophil states and assign non-canonical functions, including vascular repair and hematopoietic homeostasis. Accordingly, depletion of neutrophils compromised angiogenesis during early age, genotoxic injury, and viral infection, and impaired hematopoietic recovery after irradiation. Neutrophils acquired these properties in target tissues, a process that, in the lungs, occurred in CXCL12-rich areas and relied on CXCR4. Our results reveal that tissues co-opt neutrophils en route for elimination to induce programs that support their physiological demands.


Subject(s)
Cell Lineage , Neutrophils/metabolism , Organ Specificity , Animals , Chromatin/metabolism , Female , Hematopoiesis , Intestines/blood supply , Lung/blood supply , Male , Mice, Inbred C57BL , Neovascularization, Physiologic , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Receptors, CXCR4/metabolism , Single-Cell Analysis , Transcription, Genetic , Transcriptome/genetics
3.
Cell ; 176(5): 1128-1142.e18, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30686582

ABSTRACT

Collateral arteries are an uncommon vessel subtype that can provide alternate blood flow to preserve tissue following vascular occlusion. Some patients with heart disease develop collateral coronary arteries, and this correlates with increased survival. However, it is not known how these collaterals develop or how to stimulate them. We demonstrate that neonatal mouse hearts use a novel mechanism to build collateral arteries in response to injury. Arterial endothelial cells (ECs) migrated away from arteries along existing capillaries and reassembled into collateral arteries, which we termed "artery reassembly". Artery ECs expressed CXCR4, and following injury, capillary ECs induced its ligand, CXCL12. CXCL12 or CXCR4 deletion impaired collateral artery formation and neonatal heart regeneration. Artery reassembly was nearly absent in adults but was induced by exogenous CXCL12. Thus, understanding neonatal regenerative mechanisms can identify pathways that restore these processes in adults and identify potentially translatable therapeutic strategies for ischemic heart disease.


Subject(s)
Collateral Circulation/physiology , Heart/growth & development , Regeneration/physiology , Animals , Animals, Newborn/growth & development , Chemokine CXCL12/metabolism , Coronary Vessels/growth & development , Endothelial Cells/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/physiology , Receptors, CXCR4/metabolism , Signal Transduction
4.
Cell ; 178(5): 1088-1101.e15, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442402

ABSTRACT

Mammals evolved in the face of fluctuating food availability. How the immune system adapts to transient nutritional stress remains poorly understood. Here, we show that memory T cells collapsed in secondary lymphoid organs in the context of dietary restriction (DR) but dramatically accumulated within the bone marrow (BM), where they adopted a state associated with energy conservation. This response was coordinated by glucocorticoids and associated with a profound remodeling of the BM compartment, which included an increase in T cell homing factors, erythropoiesis, and adipogenesis. Adipocytes, as well as CXCR4-CXCL12 and S1P-S1P1R interactions, contributed to enhanced T cell accumulation in BM during DR. Memory T cell homing to BM during DR was associated with enhanced protection against infections and tumors. Together, this work uncovers a fundamental host strategy to sustain and optimize immunological memory during nutritional challenges that involved a temporal and spatial reorganization of the memory pool within "safe haven" compartments.


Subject(s)
Bone Marrow/metabolism , Immunologic Memory , Animals , Bone Marrow/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Caloric Restriction/veterinary , Cell Line, Tumor , Chemokine CXCL12/metabolism , Diet, Reducing/veterinary , Energy Metabolism , Gene Expression Regulation , Glucocorticoids , Melanoma, Experimental/mortality , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CXCR4/metabolism , Survival Rate , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , TOR Serine-Threonine Kinases/metabolism
5.
Immunity ; 57(8): 1923-1938.e7, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-38878769

ABSTRACT

Fasting is associated with improved outcomes in cancer. Here, we investigated the impact of fasting on natural killer (NK) cell anti-tumor immunity. Cyclic fasting improved immunity against solid and metastatic tumors in an NK cell-dependent manner. During fasting, NK cells underwent redistribution from peripheral tissues to the bone marrow (BM). In humans, fasting also reduced circulating NK cell numbers. NK cells in the spleen of fasted mice were metabolically rewired by elevated concentrations of fatty acids and glucocorticoids, augmenting fatty acid metabolism via increased expression of the enzyme CPT1A, and Cpt1a deletion impaired NK cell survival and function in this setting. In parallel, redistribution of NK cells to the BM during fasting required the trafficking mediators S1PR5 and CXCR4. These cells were primed by an increased pool of interleukin (IL)-12-expressing BM myeloid cells, which improved IFN-γ production. Our findings identify a link between dietary restriction and optimized innate immune responses, with the potential to enhance immunotherapy strategies.


Subject(s)
Fasting , Killer Cells, Natural , Mice, Inbred C57BL , Animals , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Humans , Neoplasms/immunology , Bone Marrow/immunology , Bone Marrow/metabolism , Mice, Knockout , Interferon-gamma/metabolism , Interferon-gamma/immunology , Spleen/immunology , Spleen/metabolism , Immunity, Innate/immunology , Interleukin-12/metabolism , Interleukin-12/immunology , Receptors, CXCR4/metabolism
6.
Nat Immunol ; 20(11): 1444-1455, 2019 11.
Article in English | MEDLINE | ID: mdl-31591573

ABSTRACT

Low exposure to microbial products, respiratory viral infections and air pollution are major risk factors for allergic asthma, yet the mechanistic links between such conditions and host susceptibility to type 2 allergic disorders remain unclear. Through the use of single-cell RNA sequencing, we characterized lung neutrophils in mice exposed to a pro-allergic low dose of lipopolysaccharide (LPS) or a protective high dose of LPS before exposure to house dust mites. Unlike exposure to a high dose of LPS, exposure to a low dose of LPS instructed recruited neutrophils to upregulate their expression of the chemokine receptor CXCR4 and to release neutrophil extracellular traps. Low-dose LPS-induced neutrophils and neutrophil extracellular traps potentiated the uptake of house dust mites by CD11b+Ly-6C+ dendritic cells and type 2 allergic airway inflammation in response to house dust mites. Neutrophil extracellular traps derived from CXCR4hi neutrophils were also needed to mediate allergic asthma triggered by infection with influenza virus or exposure to ozone. Our study indicates that apparently unrelated environmental risk factors can shape recruited lung neutrophils to promote the initiation of allergic asthma.


Subject(s)
Air Pollutants/immunology , Allergens/immunology , Asthma/immunology , Extracellular Traps/metabolism , Neutrophils/immunology , Animals , Dendritic Cells/immunology , Disease Models, Animal , Environmental Exposure/adverse effects , Extracellular Traps/immunology , Female , Humans , Lipopolysaccharides/immunology , Lung/cytology , Lung/immunology , Mice , Neutrophils/metabolism , Orthomyxoviridae/immunology , Ozone/immunology , Pyroglyphidae/immunology , Receptors, CXCR4/immunology , Receptors, CXCR4/metabolism , Up-Regulation
7.
Nat Immunol ; 20(10): 1393-1403, 2019 10.
Article in English | MEDLINE | ID: mdl-31477919

ABSTRACT

In B lymphopoiesis, activation of the pre-B cell antigen receptor (pre-BCR) is associated with both cell cycle exit and Igk recombination. Yet how the pre-BCR mediates these functions remains unclear. Here, we demonstrate that the pre-BCR initiates a feed-forward amplification loop mediated by the transcription factor interferon regulatory factor 4 and the chemokine receptor C-X-C motif chemokine receptor 4 (CXCR4). CXCR4 ligation by C-X-C motif chemokine ligand 12 activates the mitogen-activated protein kinase extracellular-signal-regulated kinase, which then directs the development of small pre- and immature B cells, including orchestrating cell cycle exit, pre-BCR repression, Igk recombination and BCR expression. In contrast, pre-BCR expression and escape from interleukin-7 have only modest effects on B cell developmental transcriptional and epigenetic programs. These data show a direct and central role for CXCR4 in orchestrating late B cell lymphopoiesis. Furthermore, in the context of previous findings, our data provide a three-receptor system sufficient to recapitulate the essential features of B lymphopoiesis in vitro.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin kappa-Chains/genetics , Precursor Cells, B-Lymphoid/physiology , Receptors, Antigen, B-Cell/metabolism , Receptors, CXCR4/metabolism , Animals , Cell Cycle Checkpoints , Cells, Cultured , Chemokine CXCL12/metabolism , Female , Interferon Regulatory Factors/genetics , Lymphopoiesis , Male , Mice , Receptors, Antigen, B-Cell/genetics , Receptors, CXCR4/genetics , Recombination, Genetic
8.
Nat Immunol ; 18(1): 15-25, 2017 01.
Article in English | MEDLINE | ID: mdl-27841869

ABSTRACT

The lymph node periphery is an important site for many immunological functions, from pathogen containment to the differentiation of helper T cells, yet the cues that position cells in this region are largely undefined. Here, through the use of a reporter for the signaling lipid S1P (sphingosine 1-phosphate), we found that cells sensed higher concentrations of S1P in the medullary cords than in the T cell zone and that the S1P transporter SPNS2 on lymphatic endothelial cells generated this gradient. Natural killer (NK) cells are located at the periphery of the lymph node, predominantly in the medulla, and we found that expression of SPNS2, expression of the S1P receptor S1PR5 on NK cells, and expression of the chemokine receptor CXCR4 were all required for NK cell localization during homeostasis and rapid production of interferon-γ by NK cells after challenge. Our findings elucidate the spatial cues for NK cell organization and reveal a previously unknown role for S1P in positioning cells within the medulla.


Subject(s)
Anion Transport Proteins/metabolism , Endothelial Cells/immunology , Killer Cells, Natural/immunology , Lymph Nodes/immunology , Lysophospholipids/metabolism , Receptors, CXCR4/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Animals , Anion Transport Proteins/genetics , Cell Differentiation , Cell Movement , Cells, Cultured , Chemotaxis , Homeostasis , Interferon-gamma/metabolism , Lymphocyte Activation/genetics , Lysophospholipids/chemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR4/genetics , Receptors, Lysosphingolipid/genetics , Signal Transduction , Sphingosine/chemistry , Sphingosine/metabolism , T-Lymphocytes, Helper-Inducer/physiology
9.
Immunity ; 51(1): 155-168.e5, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31248780

ABSTRACT

Genetic variation influences how the genome is interpreted in individuals and in mouse strains used to model immune responses. We developed approaches to utilize next-generation sequencing datasets to identify sequence variation in genes and enhancer elements in congenic and backcross mouse models. We defined genetic variation in the widely used B6-CD45.2 and B6.SJL-CD45.1 congenic model, identifying substantial differences in SJL genetic content retained in B6.SJL-CD45.1 strains on the basis of the vendor source of the mice. Genes encoding PD-1, CD62L, Bcl-2, cathepsin E, and Cxcr4 were within SJL genetic content in at least one vendor source of B6.SJL-CD45.1 mice. SJL genetic content affected enhancer elements, gene regulation, protein expression, and amino acid content in CD4+ T helper 1 cells, and mice infected with influenza showed reduced expression of Cxcr4 on B6.SJL-CD45.1 T follicular helper cells. These findings provide information on experimental variables and aid in creating approaches that account for genetic variables.


Subject(s)
Cathepsin E/metabolism , Enhancer Elements, Genetic/genetics , Immunity/genetics , Receptors, CXCR4/metabolism , Th1 Cells/immunology , Animals , Cathepsin E/genetics , Commerce , Gene Expression Regulation , Genetic Background , Genetic Variation , Germinal Center/immunology , High-Throughput Nucleotide Sequencing , Inbreeding , Leukocyte Common Antigens/genetics , Mice , Mice, Congenic , Mice, Inbred C57BL , Models, Animal , Receptors, CXCR4/genetics
10.
Immunity ; 50(2): 390-402.e10, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30709741

ABSTRACT

Neutrophils eliminate pathogens efficiently but can inflict severe damage to the host if they over-activate within blood vessels. It is unclear how immunity solves the dilemma of mounting an efficient anti-microbial defense while preserving vascular health. Here, we identify a neutrophil-intrinsic program that enabled both. The gene Bmal1 regulated expression of the chemokine CXCL2 to induce chemokine receptor CXCR2-dependent diurnal changes in the transcriptional and migratory properties of circulating neutrophils. These diurnal alterations, referred to as neutrophil aging, were antagonized by CXCR4 (C-X-C chemokine receptor type 4) and regulated the outer topology of neutrophils to favor homeostatic egress from blood vessels at night, resulting in boosted anti-microbial activity in tissues. Mice engineered for constitutive neutrophil aging became resistant to infection, but the persistence of intravascular aged neutrophils predisposed them to thrombo-inflammation and death. Thus, diurnal compartmentalization of neutrophils, driven by an internal timer, coordinates immune defense and vascular protection.


Subject(s)
Blood Vessels/immunology , Circadian Rhythm/immunology , Neutrophils/immunology , Phagocytosis/immunology , Animals , Blood Vessels/metabolism , Candida albicans/immunology , Candida albicans/physiology , Cells, Cultured , Cellular Senescence/immunology , Chemokine CXCL2/immunology , Chemokine CXCL2/metabolism , Host-Pathogen Interactions/immunology , Humans , Inflammation/immunology , Inflammation/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Receptors, CXCR4/immunology , Receptors, CXCR4/metabolism , Time Factors
11.
Cell ; 152(5): 1077-90, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23434321

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are soft tissue sarcomas that arise in connective tissue surrounding peripheral nerves. They occur sporadically in a subset of patients with neurofibromatosis type 1 (NF1). MPNSTs are highly aggressive, therapeutically resistant, and typically fatal. Using comparative transcriptome analysis, we identified CXCR4, a G-protein-coupled receptor, as highly expressed in mouse models of NF1-deficient MPNSTs, but not in nontransformed precursor cells. The chemokine receptor CXCR4 and its ligand, CXCL12, promote MPNST growth by stimulating cyclin D1 expression and cell-cycle progression through PI3-kinase (PI3K) and ß-catenin signaling. Suppression of CXCR4 activity either by shRNA or pharmacological inhibition decreases MPNST cell growth in culture and inhibits tumorigenesis in allografts and in spontaneous genetic mouse models of MPNST. We further demonstrate conservation of these activated molecular pathways in human MPNSTs. Our findings indicate a role for CXCR4 in NF1-associated MPNST development and identify a therapeutic target.


Subject(s)
Autocrine Communication , Chemokine CXCL12/metabolism , Nerve Sheath Neoplasms/metabolism , Nerve Sheath Neoplasms/pathology , Receptors, CXCR4/metabolism , Cell Cycle , Cell Proliferation , Cell Transformation, Neoplastic , Cells, Cultured , Gene Knockdown Techniques , Humans , Neurofibromatosis 1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction
12.
Immunity ; 49(3): 477-489.e7, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30231983

ABSTRACT

Adaptive immunity involves the development of bespoke antibodies in germinal centers (GCs) through immunoglobulin somatic hypermutation (SHM) in GC dark zones (DZs) and clonal selection in light zones (LZs). Accurate selection requires that cells fully replace surface B cell receptors (BCRs) following SHM, but whether this happens before LZ entry is not clear. We found that most GC B cells degrade pre-SHM receptors before leaving the DZ, and that B cells acquiring crippling mutations during SHM rarely reached the LZ. Instead, apoptosis was triggered preferentially in late G1, a stage wherein cells with functional BCRs re-entered cell cycle or reduced surface expression of the chemokine receptor CXCR4 to enable LZ migration. Ectopic expression of the anti-apoptotic gene Bcl2 was not sufficient for cells with damaging mutations to reach the LZ, suggesting that BCR-dependent cues may actively facilitate the transition. Thus, BCR replacement and pre-screening in DZs prevents the accumulation of clones with non-functional receptors and facilitates selection in the LZ.


Subject(s)
B-Lymphocytes/physiology , Clonal Selection, Antigen-Mediated , Germinal Center/immunology , Immunoglobulins/metabolism , Receptors, Antigen, B-Cell/genetics , Animals , Apoptosis , Cell Movement , Cells, Cultured , DNA Damage , Immunoglobulins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Antigen, B-Cell/metabolism , Receptors, CXCR4/metabolism , Somatic Hypermutation, Immunoglobulin
13.
PLoS Biol ; 22(4): e3002590, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38683849

ABSTRACT

Brain pericytes are one of the critical cell types that regulate endothelial barrier function and activity, thus ensuring adequate blood flow to the brain. The genetic pathways guiding undifferentiated cells into mature pericytes are not well understood. We show here that pericyte precursor populations from both neural crest and head mesoderm of zebrafish express the transcription factor nkx3.1 develop into brain pericytes. We identify the gene signature of these precursors and show that an nkx3.1-, foxf2a-, and cxcl12b-expressing pericyte precursor population is present around the basilar artery prior to artery formation and pericyte recruitment. The precursors later spread throughout the brain and differentiate to express canonical pericyte markers. Cxcl12b-Cxcr4 signaling is required for pericyte attachment and differentiation. Further, both nkx3.1 and cxcl12b are necessary and sufficient in regulating pericyte number as loss inhibits and gain increases pericyte number. Through genetic experiments, we have defined a precursor population for brain pericytes and identified genes critical for their differentiation.


Subject(s)
Brain , Pericytes , Transcription Factors , Zebrafish Proteins , Animals , Brain/metabolism , Brain/embryology , Cell Differentiation , Chemokine CXCL12/metabolism , Chemokine CXCL12/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Mesoderm/metabolism , Mesoderm/cytology , Neural Crest/metabolism , Neural Crest/cytology , Pericytes/metabolism , Pericytes/cytology , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Signal Transduction , Transcription Factors/metabolism , Transcription Factors/genetics , Zebrafish/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics
14.
Proc Natl Acad Sci U S A ; 121(14): e2304897121, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38547061

ABSTRACT

While the existence and functional role of class C G-protein-coupled receptors (GPCR) dimers is well established, there is still a lack of consensus regarding class A and B GPCR multimerization. This lack of consensus is largely due to the inherent challenges of demonstrating the presence of multimeric receptor complexes in a physiologically relevant cellular context. The C-X-C motif chemokine receptor 4 (CXCR4) is a class A GPCR that is a promising target of anticancer therapy. Here, we investigated the potential of CXCR4 to form multimeric complexes with other GPCRs and characterized the relative size of the complexes in a live-cell environment. Using a bimolecular fluorescence complementation (BiFC) assay, we identified the ß2 adrenergic receptor (ß2AR) as an interaction partner. To investigate the molecular scale details of CXCR4-ß2AR interactions, we used a time-resolved fluorescence spectroscopy method called pulsed-interleaved excitation fluorescence cross-correlation spectroscopy (PIE-FCCS). PIE-FCCS can resolve membrane protein density, diffusion, and multimerization state in live cells at physiological expression levels. We probed CXCR4 and ß2AR homo- and heteromultimerization in model cell lines and found that CXCR4 assembles into multimeric complexes larger than dimers in MDA-MB-231 human breast cancer cells and in HCC4006 human lung cancer cells. We also found that ß2AR associates with CXCR4 multimers in MDA-MB-231 and HCC4006 cells to a higher degree than in COS-7 and CHO cells and in a ligand-dependent manner. These results suggest that CXCR4-ß2AR heteromers are present in human cancer cells and that GPCR multimerization is significantly affected by the plasma membrane environment.


Subject(s)
Neoplasms , Receptors, Adrenergic, beta-2 , Receptors, CXCR4 , Signal Transduction , Animals , Cricetinae , Humans , CHO Cells , Cricetulus , Membrane Proteins/metabolism , Neoplasms/metabolism , Receptors, CXCR4/metabolism , Receptors, Adrenergic, beta-2/metabolism , Protein Multimerization
15.
PLoS Pathog ; 20(9): e1012472, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39226327

ABSTRACT

Warts, Hypogammaglobulinemia, Infections, and Myelokathexis (WHIM) syndrome is a rare primary immunodeficiency disease in humans caused by a gain of function in CXCR4, mostly due to inherited heterozygous mutations in CXCR4. One major clinical symptom of WHIM patients is their high susceptibility to human papillomavirus (HPV) induced disease, such as warts. Persistent high risk HPV infections cause 5% of all human cancers, including cervical, anogenital, head and neck and some skin cancers. WHIM mice bearing the same mutation identified in WHIM patients were created to study the underlying causes for the symptoms manifest in patients suffering from the WHIM syndrome. Using murine papillomavirus (MmuPV1) as an infection model in mice for HPV-induced disease, we demonstrate that WHIM mice are more susceptible to MmuPV1-induced warts (papillomas) compared to wild type mice. Namely, the incidence of papillomas is higher in WHIM mice compared to wild type mice when mice are exposed to low doses of MmuPV1. MmuPV1 infection facilitated both myeloid and lymphoid cell mobilization in the blood of wild type mice but not in WHIM mice. Higher incidence and larger size of papillomas in WHIM mice correlated with lower abundance of infiltrating T cells within the papillomas. Finally, we demonstrate that transplantation of bone marrow from wild type mice into WHIM mice normalized the incidence and size of papillomas, consistent with the WHIM mutation in hematopoietic cells contributing to higher susceptibility of WHIM mice to MmuPV1-induced disease. Our results provide evidence that MmuPV1 infection in WHIM mice is a powerful preclinical infectious model to investigate treatment options for alleviating papillomavirus infections in WHIM syndrome.


Subject(s)
Papillomavirus Infections , Primary Immunodeficiency Diseases , Warts , Animals , Mice , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Warts/immunology , Warts/virology , Primary Immunodeficiency Diseases/immunology , Primary Immunodeficiency Diseases/genetics , Disease Models, Animal , Papillomaviridae , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/virology , Immunologic Deficiency Syndromes/genetics , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Mice, Inbred C57BL , Disease Susceptibility , Female
16.
PLoS Pathog ; 20(8): e1012448, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39146384

ABSTRACT

The chemokine co-receptors CXCR4 and CCR5 mediate HIV entry and signal transduction necessary for viral infection. However, to date only the CCR5 antagonist maraviroc is approved for treating HIV-1 infection. Given that approximately 50% of late-stage HIV patients also develop CXCR4-tropic virus, clinical anti-HIV CXCR4 antagonists are needed. Here, we describe a novel allosteric CXCR4 antagonist TIQ-15 which inhibits CXCR4-tropic HIV-1 infection of primary and transformed CD4 T cells. TIQ-15 blocks HIV entry with an IC50 of 13 nM. TIQ-15 also inhibits SDF-1α/CXCR4-mediated cAMP production, cofilin activation, and chemotactic signaling. In addition, TIQ-15 induces CXCR4 receptor internalization without affecting the levels of the CD4 receptor, suggesting that TIQ-15 may act through a novel allosteric site on CXCR4 for blocking HIV entry. Furthermore, TIQ-15 did not inhibit VSV-G pseudotyped HIV-1 infection, demonstrating its specificity in blocking CXCR4-tropic virus entry, but not CXCR4-independent endocytosis or post-entry steps. When tested against a panel of clinical isolates, TIQ-15 showed potent inhibition against CXCR4-tropic and dual-tropic viruses, and moderate inhibition against CCR5-tropic isolates. This observation was followed by a co-dosing study with maraviroc, and TIQ-15 demonstrated synergistic activity. In summary, here we describe a novel HIV-1 entry inhibitor, TIQ-15, which potently inhibits CXCR4-tropic viruses while possessing low-level synergistic activities against CCR5-tropic viruses. TIQ-15 could potentially be co-dosed with the CCR5 inhibitor maraviroc to block viruses of mixed tropisms.


Subject(s)
HIV Infections , HIV-1 , Receptors, CXCR4 , Virus Internalization , Humans , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , HIV-1/drug effects , HIV-1/physiology , Virus Internalization/drug effects , HIV Infections/drug therapy , HIV Infections/virology , CD4-Positive T-Lymphocytes/virology , CD4-Positive T-Lymphocytes/drug effects , HIV Fusion Inhibitors/pharmacology , Maraviroc/pharmacology , Triazoles/pharmacology , Anti-HIV Agents/pharmacology , HEK293 Cells
17.
Blood ; 144(12): 1284-1289, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-38968152

ABSTRACT

ABSTRACT: Currently, the role of DNA methylation in the immunoglobulin M (IgM) monoclonal gammopathy disease spectrum remains poorly understood. In the present study, a multiomics prospective analysis was conducted integrating DNA methylation, RNA sequencing (RNA-seq), and whole-exome sequencing data in 34 subjects (23 with Waldenström macroglobulinemia [WM], 6 with IgM monoclonal gammopathy of undetermined significance [MGUS], and 5 normal controls). Analysis was focused on defining differences between IgM gammopathies (WM/IgM-MGUS) compared with controls, and specifically between WM and IgM-MGUS. Between groups, genome-wide DNA methylation analysis demonstrated a significant number of differentially methylated regions that were annotated according to genomic region. Next, integration of RNA-seq data was performed to identify potentially epigenetically deregulated pathways. We found that pathways involved in cell cycle, metabolism, cytokine/immune signaling, cytoskeleton, tumor microenvironment, and intracellular signaling were differentially activated and potentially epigenetically regulated. Importantly, there was a positive enrichment of the CXCR4 signaling pathway along with several interleukin (interleukin 6 [IL-6], IL-8, and IL-15) signaling pathways in WM compared with IgM-MGUS. Further assessment of known tumor suppressor genes and oncogenes uncovered differential promoter methylation of several targets with concordant change in gene expression, including CCND1 and CD79B. Overall, this report defines how aberrant DNA methylation in IgM gammopathies may play a critical role in the epigenetic control of oncogenesis and key cellular functions.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Immunoglobulin M , Monoclonal Gammopathy of Undetermined Significance , Waldenstrom Macroglobulinemia , Humans , Immunoglobulin M/genetics , Waldenstrom Macroglobulinemia/genetics , Waldenstrom Macroglobulinemia/immunology , Male , Monoclonal Gammopathy of Undetermined Significance/genetics , Monoclonal Gammopathy of Undetermined Significance/pathology , Monoclonal Gammopathy of Undetermined Significance/metabolism , Female , Aged , Middle Aged , Carcinogenesis/genetics , Paraproteinemias/genetics , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Prospective Studies , Signal Transduction/genetics , Multiomics
18.
Blood ; 144(9): 964-976, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-38728427

ABSTRACT

ABSTRACT: Adult hematopoietic stem and progenitor cells (HSPCs) reside in the bone marrow (BM) hematopoietic niche, which regulates HSPC quiescence, self-renewal, and commitment in a demand-adapted manner. Although the complex BM niche is responsible for adult hematopoiesis, evidence exists for simpler, albeit functional and more accessible, extramedullary hematopoietic niches. Inspired by the anecdotal description of retroperitoneal hematopoietic masses occurring at higher frequency upon hormonal dysregulation within the adrenal gland, we hypothesized that the adult adrenal gland could be induced into a hematopoietic-supportive environment in a systematic manner, thus revealing mechanisms underlying de novo niche formation in the adult. Here, we show that upon splenectomy and hormonal stimulation, the adult adrenal gland of mice can be induced to recruit and host functional HSPCs, capable of serial transplantation, and that this phenomenon is associated with de novo formation of platelet-derived growth factor receptor α/leptin receptor (PDGFRα+/LEPR+/-)-expressing stromal nodules. We further show in CXCL12-green fluorescent protein reporter mice that adrenal glands contain a stromal population reminiscent of the CXCL12-abundant reticular cells, which compose the BM HSPC niche. Mechanistically, HSPC homing to hormonally induced adrenal glands was found dependent on the CXCR4-CXCL12 axis. Mirroring our findings in mice, we found reticular CXCL12+ cells coexpressing master niche regulator FOXC1 in primary samples from human adrenal myelolipomas, a benign tumor composed of adipose and hematopoietic tissue. Our findings reignite long-standing questions regarding hormonal regulation of hematopoiesis and provide a novel model to facilitate the study of adult-specific inducible hematopoietic niches, which may pave the way to therapeutic applications.


Subject(s)
Adrenal Glands , Chemokine CXCL12 , Hematopoietic Stem Cells , Receptors, CXCR4 , Stem Cell Niche , Animals , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Chemokine CXCL12/metabolism , Mice , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Adrenal Glands/metabolism , Adrenal Glands/pathology , Mice, Inbred C57BL , Humans , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptors, Leptin/metabolism , Receptors, Leptin/genetics , Hematopoiesis, Extramedullary , Splenectomy , Mice, Transgenic
19.
J Immunol ; 212(7): 1075-1080, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38363205

ABSTRACT

B cell trafficking involves the coordinated activity of multiple adhesive and cytokine-receptor interactions, and the players in this process are not fully understood. In this study, we identified the tetraspanin CD53 as a critical regulator of both normal and malignant B cell trafficking. CXCL12 is a key chemokine in B cell homing to the bone marrow and secondary lymphoid organs, and both normal and malignant B cells from Cd53-/- mice have reduced migration toward CXCL12 in vitro, as well as impaired marrow homing in vivo. Using proximity ligation studies, we identified the CXCL12 receptor, CXCR4, as a novel, to our knowledge, CD53 binding partner. This interaction promotes receptor function, because Cd53-/- B cells display reduced signaling and internalization of CXCR4 in response to CXCL12. Together, our data suggest that CD53 interacts with CXCR4 on both normal and malignant B cells to promote CXCL12 signaling, receptor internalization, and marrow homing.


Subject(s)
B-Lymphocytes , Bone Marrow , Animals , Mice , Bone Marrow/metabolism , B-Lymphocytes/metabolism , Chemokine CXCL12/metabolism , Signal Transduction , Tetraspanins/metabolism , Carrier Proteins/metabolism , Receptors, CXCR4/metabolism , Cell Movement/physiology , Bone Marrow Cells/metabolism
20.
Mol Cell ; 70(1): 106-119.e10, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29625032

ABSTRACT

A current challenge in cell motility studies is to understand the molecular and physical mechanisms that govern chemokine receptor nanoscale organization at the cell membrane, and their influence on cell response. Using single-particle tracking and super-resolution microscopy, we found that the chemokine receptor CXCR4 forms basal nanoclusters in resting T cells, whose extent, dynamics, and signaling strength are modulated by the orchestrated action of the actin cytoskeleton, the co-receptor CD4, and its ligand CXCL12. We identified three CXCR4 structural residues that are crucial for nanoclustering and generated an oligomerization-defective mutant that dimerized but did not form nanoclusters in response to CXCL12, which severely impaired signaling. Overall, our data provide new insights to the field of chemokine biology by showing that receptor dimerization in the absence of nanoclustering is unable to fully support CXCL12-mediated responses, including signaling and cell function in vivo.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Membrane/metabolism , Cell Movement , Nanoparticles , Receptors, CXCR4/metabolism , T-Lymphocytes/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/immunology , Amino Acid Motifs , Animals , CD4 Antigens/metabolism , Cell Membrane/drug effects , Cell Membrane/immunology , Chemokine CXCL12/pharmacology , HEK293 Cells , Humans , Jurkat Cells , Ligands , Mice, Inbred C57BL , Mutation , Protein Multimerization , Protein Transport , Receptors, CXCR4/drug effects , Receptors, CXCR4/genetics , Receptors, CXCR4/immunology , Signal Transduction , Single Molecule Imaging , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL