Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 182
1.
Clin Nucl Med ; 49(6): 592-593, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38630995

ABSTRACT: An 83-year-old woman with newly diagnosed multiple myeloma (MM) was enrolled in our 68 Ga-pentixather and 68 Ga-pentixafor PET/CT trial for evaluation of tumor burden. 68 Ga-pentixather PET/CT detected more focal bone lesions, and the uptake levels of focal bone lesions on 68 Ga-pentixather PET/CT were higher than those on 68 Ga-pentixafor PET/CT. This suggests that 68 Ga-pentixather PET/CT may be an alternative imaging modality and more sensitive in detecting MM lesions than 68 Ga-pentixafor PET/CT.


Multiple Myeloma , Positron Emission Tomography Computed Tomography , Aged, 80 and over , Female , Humans , Coordination Complexes , Gallium Radioisotopes , Multiple Myeloma/diagnostic imaging , Peptides, Cyclic , Receptors, CXCR4/drug effects
2.
Drug Des Devel Ther ; 16: 67-81, 2022.
Article En | MEDLINE | ID: mdl-35023903

BACKGROUND: Cross-sensitization of pelvic organs is one theory for why symptoms of gut sickness and interstitial cystitis/bladder pain syndrome overlap. Experimental colitis has been shown to trigger bladder hyperactivity and hyperalgesia in rats. The chemokine receptor CXCR4 plays a key role in bladder function and central sensitization. We aim to study the role of CXCR4 and its inhibitor AMD3100 in colon-bladder cross-organ sensitization. METHODS: The colitis model was established by rectal infusion of trinitrobenzene sulfonic acid. Western blot and immunofluorescence were used to assess the expression and distribution of CXCR4. Intrathecal injection of AMD3100 (a CXCR4 inhibitor) and PD98059 (an ERK inhibitor) were used to inhibit CXCR4 and downstream extracellular signal-regulated kinase (ERK) in the spinal cord and dorsal root ganglion (DRG). Intravesical perfusion of resiniferatoxin was performed to measure the pain behavior counts of rats, and continuous cystometry was performed to evaluate bladder voiding function. RESULTS: Compared to the control group, CXCR4 was expressed more in bladder mucosa and colon mucosa, L6-S1 dorsal root ganglion (DRG), and the corresponding segment of the spinal dorsal horn (SDH) in rats with colitis. Moreover, intrathecal injection of the AMD3100 suppressed bladder overactivity, bladder hyperalgesia, and mastocytosis symptoms caused by colitis. Furthermore, AMD3100 effectively inhibited ERK activation in the spinal cord induced by experimental colitis. Finally, treatment with PD98059 alleviated bladder overactivity and hyperalgesia caused by colitis. CONCLUSION: Increased CXCR4 in the DRG and SDH contributes to colon inflammation-induced bladder overactivity and hyperalgesia partly via the phosphorylation of spinal ERK. Treatment targeting the CXCR4/ERK pathway might provide a potential new approach for the comorbidity between the digestive system and the urinary system.


Benzylamines/pharmacology , Colitis/drug therapy , Colitis/metabolism , Cyclams/pharmacology , Ganglia, Spinal/metabolism , Receptors, CXCR4/drug effects , Spinal Cord/metabolism , Animals , Benzylamines/administration & dosage , Colitis/complications , Cyclams/administration & dosage , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Flavonoids/administration & dosage , Flavonoids/pharmacology , Hyperalgesia/chemically induced , Pain Measurement , Rats , Rats, Sprague-Dawley , Receptors, CXCR4/metabolism , Signal Transduction , Urinary Bladder Diseases/drug therapy , Urinary Bladder Diseases/etiology , Urinary Bladder Diseases/metabolism , Urination/drug effects
4.
Immunohorizons ; 5(12): 994-1007, 2021 12 29.
Article En | MEDLINE | ID: mdl-34965966

Monocytes and macrophages are early sentinels of infection. The peritoneum contains two resident populations: large and small peritoneal macrophages (LPMs and SPMs). While LPMs self-renew, circulating monocytes enter the peritoneum and differentiate into SPMs. We lack information on the dynamics of monocyte-macrophage trafficking during abdominal sepsis, reflecting an important knowledge gap. In this study, we characterize the presence of LPMs, SPMs, and monocytes in the peritoneum of mice following cecal ligation and puncture (CLP)-induced sepsis and sham surgery. LPMs rapidly disappeared from the peritoneum and were scarce at 18-66 h after CLP or sham surgery. By 14 d, LPMs returned for sham mice, but they remained scarce in CLP mice. Depletion of LPMs from the peritoneum of CD11b-DTR mice greatly increased animal mortality. These data imply that LPMs are critical for sepsis survival. Monocytes rapidly infiltrated the peritoneum and were abundant at 18-66 h after CLP or sham surgery. Surprisingly, SPMs only increased at 14 d post-CLP. Therefore, monocytes may defend hosts from acute sepsis mortality without generating SPMs. More monocytes were present in mice predicted to survive sepsis versus mice predicted to die. However, altering monocyte numbers via CCR2 deficiency or adoptive transfer did not significantly affect animal survival. We reasoned that animals destined to survive sepsis may exhibit a different monocyte phenotype, rather than merely enhanced numbers. Indeed, mice predicted to survive possessed more CD31+, CXCR4hi transitional premonocytes in their abdomen. Inhibition of CXCL12-CXCR4 signaling via AMD3100 exacerbated sepsis. These data imply that recruitment of transitional premonocytes to the abdomen promotes sepsis survival.


Macrophages, Peritoneal/pathology , Sepsis/mortality , Sepsis/pathology , Animals , Benzylamines/pharmacology , Chemokine CXCL12/drug effects , Cyclams/pharmacology , Disease Models, Animal , Female , Ligation , Macrophages/metabolism , Macrophages, Peritoneal/immunology , Male , Mice , Monocytes/metabolism , Receptors, CXCR4/drug effects , Sepsis/drug therapy , Sepsis/immunology
5.
Pharmacol Res Perspect ; 9(5): e00873, 2021 10.
Article En | MEDLINE | ID: mdl-34632734

We hypothesized whether propofol or active propofol component (2,6-diisopropylphenol [DIPPH] and lipid excipient [LIP-EXC]) separately may alter inflammatory mediators expressed by macrophages and neutrophils in lean and obese rats. Male Wistar rats (n = 10) were randomly assigned to receive a standard (lean) or obesity-inducing diet (obese) for 12 weeks. Animals were euthanized, and alveolar macrophages and neutrophils from lean and obese animals were exposed to propofol (50 µM), active propofol component (50 µM, 2,6-DIPPH), and lipid excipient (soybean oil, purified egg phospholipid, and glycerol) for 1 h. The primary outcome was IL-6 expression after propofol and its components exposure by alveolar macrophages extracted from bronchoalveolar lavage fluid. The secondary outcomes were the production of mediators released by macrophages from adipose tissue, and neutrophils from lung and adipose tissues, and neutrophil migration. IL-6 increased after the exposure to both propofol (median [interquartile range] 4.14[1.95-5.20]; p = .04) and its active component (2,6-DIPPH) (4.09[1.67-5.91]; p = .04) in alveolar macrophages from obese animals. However, only 2,6-DIPPH increased IL-10 expression (7.59[6.28-12.95]; p = .001) in adipose tissue-derived macrophages. Additionally, 2,6-DIPPH increased C-X-C chemokine receptor 2 and 4 (CXCR2 and CXCR4, respectively) in lung (10.08[8.23-29.01]; p = .02; 1.55[1.49-3.43]; p = .02) and adipose tissues (8.78[4.15-11.57]; p = .03; 2.86[2.17-3.71]; p = .01), as well as improved lung-derived neutrophil migration (28.00[-3.42 to 45.07]; p = .001). In obesity, the active component of propofol affected both the M1 and M2 markers as well as neutrophils in both alveolar and adipose tissue cells, suggesting that lipid excipient may hinder the effects of active propofol.


Adipose Tissue/drug effects , Anesthetics, Intravenous/pharmacology , Excipients/pharmacology , Interleukin-6/metabolism , Lung/drug effects , Macrophages, Alveolar/drug effects , Neutrophils/drug effects , Obesity/metabolism , Propofol/pharmacology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Chemotaxis, Leukocyte/drug effects , Glycerol/pharmacology , Interleukin-10/metabolism , Lung/metabolism , Macrophages, Alveolar/metabolism , Neutrophils/metabolism , Phospholipids/pharmacology , Rats , Receptors, CXCR4/drug effects , Receptors, CXCR4/metabolism , Receptors, Interleukin-8B/drug effects , Receptors, Interleukin-8B/metabolism , Soybean Oil/pharmacology
6.
Commun Biol ; 4(1): 569, 2021 05 12.
Article En | MEDLINE | ID: mdl-33980979

Following the FDA-approval of the hematopoietic stem cell (HSC) mobilizer plerixafor, orally available and potent CXCR4 antagonists were pursued. One such proposition was AMD11070, which was orally active and had superior antagonism in vitro; however, it did not appear as effective for HSC mobilization in vivo. Here we show that while AMD11070 acts as a full antagonist, plerixafor acts biased by stimulating ß-arrestin recruitment while fully antagonizing G protein. Consequently, while AMD11070 prevents the constitutive receptor internalization, plerixafor allows it and thereby decreases receptor expression. These findings are confirmed by the successful transfer of both ligands' binding sites and action to the related CXCR3 receptor. In vivo, plerixafor exhibits superior HSC mobilization associated with a dramatic reversal of the CXCL12 gradient across the bone marrow endothelium, which is not seen for AMD11070. We propose that the biased action of plerixafor is central for its superior therapeutic effect in HSC mobilization.


Benzylamines/pharmacology , Cyclams/pharmacology , Hematopoietic Stem Cell Mobilization/methods , Receptors, CXCR4/metabolism , Aminoquinolines/metabolism , Aminoquinolines/pharmacology , Animals , Benzimidazoles/metabolism , Benzimidazoles/pharmacology , Benzylamines/metabolism , Butylamines/metabolism , Butylamines/pharmacology , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Cyclams/metabolism , Drug Delivery Systems/methods , Female , Granulocyte Colony-Stimulating Factor , HEK293 Cells , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Pharmaceutical Preparations/metabolism , Receptors, CXCR3/drug effects , Receptors, CXCR3/metabolism , Receptors, CXCR4/drug effects , beta-Arrestins/drug effects , beta-Arrestins/metabolism
7.
Curr Pharm Biotechnol ; 22(8): 1122-1128, 2021.
Article En | MEDLINE | ID: mdl-33023442

BACKGROUND: Aloe vera has been reported as a topical antibiotic and healing agent for wounds, but advantages of oral administration and mechanisms of wound healing have not been reported. Present study focuses on the evaluation of effects of oral administration of Aloe vera for excisional cutaneous wounds in Sprague Dawley rats. METHODS: Sprague Dawley (SD) rats were inflicted with excisional wounds and were either treated with Aloe vera orally (Aloe vera) or kept untreated (wound). In contrast, healthy rats were kept as control group. Wound area was measured from day 7th to day 21st. Collagen content was estimated by hydroxyproline assay. Histology was analysed by hematoxylin and eosin staining. Angiogenesis was observed by indirect ELISA for Insulin like Growth Factor (IGF-1) and Vascular Endothelial Growth Factor (VEGF) protein from skin, serum and bone marrow. Chemotaxis was evaluated by RT-PCR analysis for Stromal cell-Derived Factor-1 (SDF-1) and C-X-C chemokine receptor type 4 (CXCR-4) from skin and bone marrow. RESULTS: Aloe vera healed wounds earlier than untreated rats with gradual improvement in wound areas and collagen content. Aloe vera also improved the expression of IGF-1 and VEGF in skin and bone marrow indicating an improvement in angiogenesis. RT- PCR analysis showed increased expression of genes for chemotaxis (SDF-1 and CXCR-4) in skin and bone marrow. CONCLUSION: Aloe vera improves healing by increasing collagen content, improving angiogenesis and chemotaxis.


Aloe , Neovascularization, Physiologic/drug effects , Plant Extracts/pharmacology , Wound Healing/drug effects , Administration, Oral , Animals , Chemokine CXCL12/drug effects , Chemotaxis/drug effects , Collagen/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, CXCR4/drug effects , Skin/drug effects , Somatomedins/drug effects , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/metabolism
8.
Proc Natl Acad Sci U S A ; 117(46): 28960-28970, 2020 11 17.
Article En | MEDLINE | ID: mdl-33127761

Inhibition of the chemokine receptor CXCR4 in combination with blockade of the PD-1/PD-L1 T cell checkpoint induces T cell infiltration and anticancer responses in murine and human pancreatic cancer. Here we elucidate the mechanism by which CXCR4 inhibition affects the tumor immune microenvironment. In human immune cell-based chemotaxis assays, we find that CXCL12-stimulated CXCR4 inhibits the directed migration mediated by CXCR1, CXCR3, CXCR5, CXCR6, and CCR2, respectively, chemokine receptors expressed by all of the immune cell types that participate in an integrated immune response. Inhibiting CXCR4 in an experimental cancer medicine study by 1-wk continuous infusion of the small-molecule inhibitor AMD3100 (plerixafor) induces an integrated immune response that is detected by transcriptional analysis of paired biopsies of metastases from patients with microsatellite stable colorectal and pancreatic cancer. This integrated immune response occurs in three other examples of immune-mediated damage to noninfected tissues: Rejecting renal allografts, melanomas clinically responding to anti-PD1 antibody therapy, and microsatellite instable colorectal cancers. Thus, signaling by CXCR4 causes immune suppression in human pancreatic ductal adenocarcinoma and colorectal cancer by impairing the function of the chemokine receptors that mediate the intratumoral accumulation of immune cells.


Colorectal Neoplasms/metabolism , Immunity/immunology , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Receptors, CXCR4/drug effects , Receptors, CXCR4/metabolism , Aged , Benzylamines , Carcinoma, Pancreatic Ductal , Chemokine CXCL12 , Colorectal Neoplasms/pathology , Cyclams , Female , Heterocyclic Compounds/antagonists & inhibitors , Humans , Immunotherapy , Male , Middle Aged , Pancreatic Neoplasms/pathology , Receptors, CCR2/metabolism , Receptors, CXCR3/metabolism , Receptors, CXCR5/metabolism , Receptors, CXCR6/metabolism , Receptors, Interleukin-8A/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/immunology , Pancreatic Neoplasms
9.
Alcohol Clin Exp Res ; 44(4): 866-879, 2020 04.
Article En | MEDLINE | ID: mdl-32020622

BACKGROUND: Prenatal exposure to ethanol (EtOH) has lasting effects on neuropeptide and neuroimmune systems in the brain alongside detrimental alcohol-related behaviors. At low-to-moderate doses, prenatal EtOH stimulates neurogenesis in lateral hypothalamus (LH) and increases neurons that express the orexigenic peptides hypocretin/orexin (Hcrt/OX) and melanin-concentrating hormone (MCH), and the proinflammatory chemokine CCL2, which through its receptor CCR2 stimulates cell differentiation and movement. Our recent studies demonstrated that CCL2 and CCR2 colocalize with MCH neurons and are involved in EtOH's stimulatory effect on their development but show no relation to Hcrt/OX. Here, we investigated another chemokine, CXCL12, and its receptor, CXCR4, which promote neurogenesis and neuroprogenitor cell proliferation, to determine if they also exhibit peptide specificity in their response to EtOH exposure. METHODS: Pregnant rats were intraorally administered a moderate dose of EtOH (2 g/kg/d) from embryonic day 10 (E10) to E15. Their embryos and postnatal offspring were examined using real-time quantitative PCR and immunofluorescence histochemistry, to determine if EtOH affects CXCL12 and CXCR4 and the colocalization of CXCR4 with Hcrt/OX and MCH neurons in the LH and with radial glia neuroprogenitor cells in the hypothalamic neuroepithelium (NEP). RESULTS: Prenatal EtOH strongly stimulated CXCL12 and CXCR4 in LH neurons of embryos and postnatal offspring. This stimulation was significantly stronger in Hcrt/OX than MCH neurons in LH and also occurred in radial glia neuroprogenitor cells dense in the NEP. These effects were sexually dimorphic, consistently stronger in females than males. CONCLUSIONS: While showing prenatal EtOH exposure to have a sexually dimorphic, stimulatory effect on CXCL12 and CXCR4 in LH similar to CCL2 and its receptor, these results reveal their distinct relationship to the peptide neurons, with the former closely related to Hcrt/OX and the latter to MCH, and they link EtOH's actions in LH to a stimulatory effect on neuroprogenitor cells in the NEP.


Central Nervous System Depressants/pharmacology , Chemokine CXCL12/drug effects , Ependymoglial Cells/drug effects , Ethanol/pharmacology , Hypothalamic Area, Lateral/drug effects , Neural Stem Cells/drug effects , Neurons/drug effects , Receptors, CXCR4/drug effects , Animals , Animals, Newborn , Cell Proliferation/drug effects , Chemokine CXCL12/metabolism , Embryo, Mammalian , Ependymoglial Cells/metabolism , Hypothalamic Area, Lateral/cytology , Hypothalamic Area, Lateral/metabolism , Hypothalamic Hormones/metabolism , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus/metabolism , Immunohistochemistry , Melanins/metabolism , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurons/metabolism , Orexins/metabolism , Pituitary Hormones/metabolism , Rats , Real-Time Polymerase Chain Reaction , Receptors, CXCR4/metabolism
10.
Expert Opin Ther Pat ; 30(2): 87-101, 2020 Feb.
Article En | MEDLINE | ID: mdl-31854208

Introduction: The chemokine receptor CXCR4 has been under intense study due to the central role it plays in immune system regulation and the pathology of many human diseases. The FDA approval of the first CXCR4 antagonist drug Plerixafor (i.e. AMD3100, Mozobil®) ushered in an increase in patent activity covering CXCR4 based therapeutic agents over the past decade.Areas covered: This article describes patent documents published during the period of 2010 through 2018 for both small molecules and peptide-based CXCR4 modulators as therapeutic agents. There is an expansion of intellectual property (IP) around existing and new small molecules of clinical interest, including new chemotypes featuring aromatic and aliphatic heterocycles. There is also significant IP covering peptide-based therapeutics, although about half as many in number as those covering small molecules.Expert opinion: In the last decade there has been significant interest in modulators of the CXCR4 receptor, as gauged by the number of patent filings and clinical investigations targeting this receptor for human disease intervention. Seven of the many CXCR4 modulators described herein, that are currently in human clinical trials, are likely to spur the creation of other FDA approved therapeutics in the near future, most likely as immune and oncology drugs.


Drug Development , Peptides/pharmacology , Receptors, CXCR4/drug effects , Animals , Benzylamines , Cyclams , Heterocyclic Compounds/pharmacology , Humans , Patents as Topic , Receptors, CXCR4/metabolism
11.
Medicina (Kaunas) ; 55(6)2019 Jun 21.
Article En | MEDLINE | ID: mdl-31234437

Background and objectives: To enter the target cell, HIV-1 binds not only CD4 but also a co-receptor ß-chemokine receptor 5 (CCR5) or α chemokine receptor 4 (CXCR4). Limited information is available on the impact of co-receptor usage on HIV-1 replication in monocyte-derived macrophages (MDM) and on the homeostasis of this important cellular reservoir. Materials and Methods: Replication (measured by p24 production) of the CCR5-tropic 81A strain increased up to 10 days post-infection and then reached a plateau. Conversely, the replication of the CXCR4-tropic NL4.3 strain (after an initial increase up to day 7) underwent a drastic decrease becoming almost undetectable after 10 days post-infection. The ability of CCR5-tropic and CXCR4-tropic strains to induce cell death in MDM was then evaluated. While for CCR5-tropic 81A the rate of apoptosis in MDM was comparable to uninfected MDM, the infection of CXCR4-tropic NL4.3 in MDM was associated with a rate of 14.3% of apoptotic cells at day 6 reaching a peak of 43.5% at day 10 post-infection. Results: This suggests that the decrease in CXCR4-tropic strain replication in MDM can be due to their ability to induce cell death in MDM. The increase in apoptosis was paralleled with a 2-fold increase in the phosphorylated form of p38 compared to WT. Furthermore, microarray analysis showed modulation of proapoptotic and cancer-related genes induced by CXCR4-tropic strains starting from 24 h after infection, whereas CCR5 viruses modulated the expression of genes not correlated with apoptotic-pathways. Conclusions: In conclusion, CXCR4-tropic strains can induce a remarkable depletion of MDM. Conversely, MDM can represent an important cellular reservoir for CCR5-tropic strains supporting the role of CCR5-usage in HIV-1 pathogenesis and as a pharmacological target to contribute to an HIV-1 cure.


HIV-1/drug effects , HIV-1/growth & development , Heterocyclic Compounds/pharmacology , Macrophages/drug effects , Anti-HIV Agents/pharmacology , Benzylamines , Cyclams , DNA Fragmentation/drug effects , HIV-1/isolation & purification , Humans , Receptors, CCR5/drug effects , Receptors, CCR5/genetics , Receptors, CXCR4/drug effects , Receptors, CXCR4/genetics
12.
J Endod ; 45(5): 599-605.e1, 2019 May.
Article En | MEDLINE | ID: mdl-30926162

INTRODUCTION: In regenerative endodontics, irrigation is an important step to ensure the success of treatment. EDTA as a common irrigant has been recommended in the American Associations of Endodontists guidelines. It has been suggested that EDTA-treated dentin slices could increase the attachment, differentiation, and migration of dental pulp stem cells. However, no information is available about the effect of EDTA on the migration of dental pulp cells (DPCs). The aim of this study was to explore how EDTA affects the migration of DPCs. METHODS: Cells were obtained from human premolars or third molars, and cell counting kit-8 was used to evaluate the influence of EDTA on cell proliferation at various concentrations and time points. Real-time polymerase chain reaction was used to detect the messenger RNA expression levels of transforming growth factor beta (TGF-ß) and chemokine receptor 4 (CXCR4). Protein expression was tested by the enzyme-linked immunosorbent assay and Western blot, respectively. In addition, the transwell migration assay was performed to investigate the role of EDTA pretreatment in stromal cell-derived factor 1α (SDF-1α)-induced DPC migration. RESULTS: Stimulation with 12% EDTA enhanced SDF-1α-induced migration of DPCs. Both expressions of TGF-ß1 and CXCR4 were increased by 12% EDTA in a time-dependent manner. After silencing CXCR4, EDTA-enhanced migration was decreased. Furthermore, the transcriptional regulation of CXCR4 by EDTA was found to be mediated by TGF-ß1/ERK1/2 and TGF-ß1/Smad2/3 signal pathways. CONCLUSIONS: Our results showed that 12% EDTA could promote SDF-1α-induced migration of DPCs by up-regulating CXCR4 expression in which TGF-ß1 signal pathways were involved.


Calcium Chelating Agents , Cell Movement , Chemokine CXCL12 , Dental Pulp , Edetic Acid , Receptors, CXCR4 , Stromal Cells , Calcium Chelating Agents/pharmacology , Cell Movement/drug effects , Edetic Acid/pharmacology , Humans , Receptors, CXCR4/drug effects , Receptors, CXCR4/metabolism , Signal Transduction , Stromal Cells/drug effects
13.
Cell Biol Int ; 42(10): 1321-1329, 2018 Sep.
Article En | MEDLINE | ID: mdl-29908007

Mesenchymal stem cells homing and migration is a crucial step during bone fracture healing. Hypoxic environment in fracture site induces bone marrow mesenchymal stem cells (BMSCs) migration, but its mechanism remains unclear. Our previous study and studies by other groups have reported the involvement of signal transducer and activator of transcription 3 (STAT3) pathway in cell migration. However, the role of STAT3 pathway in hypoxia-induced cell migration is still unknown. In this study, we investigated the role of STAT3 signaling in hypoxia-induced BMSCs migration and osteogenic differentiation. BMSCs isolated from C57BL/6 male mice were cultured in the presence of cobalt chloride (CoCl2 ) to simulate intracellular hypoxia. Hypoxia enhanced BMSCs migration, and upregulated cell migration related gene expression, that is, metalloproteinase (MMP) 7, MMP9, and C-X-C motif chemokine receptor 4. Hypoxia enhanced the phosphorylation of STAT3, and cell migration related proteins: c-jun n-terminal kinase (JNK), focal of adhesion kinase (FAK), extracellular regulated protein kinases, and protein kinase B 1/2 (ERK1/2). Moreover, hypoxia enhanced expression of osteogenic differentiation marker. Inhibition of STAT3 suppressed the hypoxia-induced BMSCs migration, cell migration related signaling molecules phosphorylation, and osteogenic differentiation related gene expression. In conclusion, our result indicates that hypoxia-induced BMSCs migration and osteogenic differentiation is via STAT3 phosphorylation and involves the cooperative activity of the JNK, FAK, and MMP9 signaling pathways.


Mesenchymal Stem Cells/drug effects , STAT3 Transcription Factor/drug effects , Animals , Bone Marrow Cells/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cobalt/pharmacology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Expression Regulation/drug effects , Hypoxia/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Matrix Metalloproteinase 7/drug effects , Matrix Metalloproteinase 9/drug effects , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Osteogenesis/genetics , Phosphorylation , Receptors, CXCR4/drug effects , Signal Transduction/drug effects
14.
Mol Cell ; 70(1): 106-119.e10, 2018 04 05.
Article En | MEDLINE | ID: mdl-29625032

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.


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
15.
ACS Chem Biol ; 13(4): 881-886, 2018 04 20.
Article En | MEDLINE | ID: mdl-29461034

CXC chemokine receptor 4 (CXCR4) is involved in multiple physiological and pathological processes, notably as a coreceptor for human immunodeficiency virus (HIV) cell entry. Its broad expression pattern and vital biological importance make CXCR4 a troublesome drug target, as disruption of the interaction with its endogenous ligand, CXC chemokine ligand 12 (CXCL12), has severe consequences. In fact, only one CXCR4 drug, the bicyclam antagonist and HIV entry inhibitor AMD3100 (Plerixafor/Mozobil), has been approved for clinical use, however only for stem cell mobilization-a consequence of CXCR4 antagonism. Here, we report the engineering of an efficacy switch mutation in CXCR4-F292A7.43 in the middle of transmembrane helix 7-that converted the antagonists AMD3100 and AMD11070 into partial agonists. As agonists on F292A CXCR4, AMD3100 and AMD11070 were less disruptive to CXCR4 signaling while they remained efficient inhibitors of HIV fusion. This demonstrates that small molecule CXCR4 agonists can have a therapeutic potential as HIV entry inhibitors.


HIV Fusion Inhibitors/chemistry , HIV-1/drug effects , Receptors, CXCR4/drug effects , Anti-HIV Agents , Humans , Protein Engineering/methods , Receptors, CXCR4/agonists , Receptors, CXCR4/antagonists & inhibitors
16.
Shanghai Kou Qiang Yi Xue ; 27(6): 579-584, 2018 Dec.
Article Zh | MEDLINE | ID: mdl-30899936

PURPOSE: To evaluate the effect of different concentrations of rhPDGF-BB on bone marrow stromal stem cells(BMSCs) migration in diabetic rat, and related mechanisms of SDF-1 and its G-protein conjugated receptor regulation, provide a theoretical basis for the application of rhPDGF-BB to diabetic patients for bone regeneration therapy. METHODS: Animal model with diabetes were induced by streptozotocin in rats. BMSCs of diabetic rats were cultured in vitro as the control group. Transwell chemokinesis model was adopted to detect the chemokinesis of BMSCs in vitro with rhPDGF-BB at concentrations of 0, 10, 50, and 100 ng/mL, and real-time PCR was used to detect the expression of BMSCs SDF-1, CXCR4 mRNA, determine the optimum concentration of the drug. The regulatory effects of rhPDGF-BB on SDF-1 and CXCR4 expression of BMSCs were confirmed in reverse with PI3K/Akt inhibitor. Statistical analysis of the data was performed using SPSS 17.0 software package. RESULTS: In diabetic rats induced by streptozotocin, one week later, the rat tail vein blood glucose concentration higher than 16.7 mmol/L was selected as the successful model rats. Compared with BMSCs in diabetic rats, rhPDGF-BB promoted BMSCs migration in diabetic rats, 50 ng/mL rhPDGF-BB was the optimal concentration in diabetic rats, and PI3K/Akt inhibitors significantly inhibited the migration of BMSCs in diabetic rats. CONCLUSIONS: rhPDGF-BB promotes the migration of diabetic rat BMSCs and regulates its migration by regulating SDF-1/CXCR4 axis.


Becaplermin , Bone Marrow Cells , Diabetes Mellitus, Experimental , Mesenchymal Stem Cells , Animals , Becaplermin/pharmacology , Bone Marrow Cells/physiology , Bone Regeneration , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , Phosphatidylinositol 3-Kinases , Rats , Rats, Sprague-Dawley , Receptors, CXCR4/drug effects , Receptors, CXCR4/metabolism , Recombinant Proteins
17.
Cell Host Microbe ; 22(1): 99-110.e7, 2017 Jul 12.
Article En | MEDLINE | ID: mdl-28704658

HIV-1 entry into host cells starts with interactions between the viral envelope glycoprotein (Env) and cellular CD4 receptors and coreceptors. Previous work has suggested that efficient HIV entry also depends on intracellular signaling, but this remains controversial. Here we report that formation of the pre-fusion Env-CD4-coreceptor complexes triggers non-apoptotic cell surface exposure of the membrane lipid phosphatidylserine (PS). HIV-1-induced PS redistribution depends on Ca2+ signaling triggered by Env-coreceptor interactions and involves the lipid scramblase TMEM16F. Externalized PS strongly promotes Env-mediated membrane fusion and HIV-1 infection. Blocking externalized PS or suppressing TMEM16F inhibited Env-mediated fusion. Exogenously added PS promoted fusion, with fusion dependence on PS being especially strong for cells with low surface density of coreceptors. These findings suggest that cell-surface PS acts as an important cofactor that promotes the fusogenic restructuring of pre-fusion complexes and likely focuses the infection on cells conducive to PS signaling.


HIV Infections/virology , HIV-1/physiology , HIV-1/pathogenicity , Membrane Fusion/physiology , Phosphatidylserines/metabolism , Virus Activation/physiology , Virus Internalization , Amides/antagonists & inhibitors , Anoctamins/metabolism , Antibodies, Monoclonal , Benzylamines , CD4 Antigens/metabolism , Calcium/metabolism , Cell Line , Cell Membrane/metabolism , Cyclams , HEK293 Cells , HeLa Cells , Heterocyclic Compounds/antagonists & inhibitors , Host-Pathogen Interactions/physiology , Humans , Phospholipid Transfer Proteins/metabolism , Quaternary Ammonium Compounds/antagonists & inhibitors , Receptors, CCR5/drug effects , Receptors, CCR5/immunology , Receptors, CXCR4/drug effects , Signal Transduction , Viral Envelope Proteins/metabolism , Virus Attachment , Virus Replication/physiology
18.
Nat Med ; 23(4): 424-428, 2017 Apr.
Article En | MEDLINE | ID: mdl-28263313

Efficient hematopoietic stem cell (HSC) homing is important for hematopoietic cell transplantation (HCT), especially when HSC numbers are limited, as in the use of cord blood (CB). In a screen of small-molecule compounds, we identified glucocorticoid (GC) hormone signaling as an activator of CXCR4 expression in human CB HSCs and hematopoietic progenitor cells (HPCs). Short-term GC pretreatment of human CB HSCs and HPCs promoted SDF-1-CXCR4-axis-mediated chemotaxis, homing, and long-term engraftment when these cells were transplanted into primary- and secondary-recipient NSG mice. Mechanistically, activated glucocorticoid receptor binds directly to a glucocorticoid response element in the CXCR4 promoter and recruits the SRC-1-p300 complex to promote H4K5 and H4K16 histone acetylation, facilitating transcription of CXCR4. These results suggest a new and readily available means to enhance the clinical efficacy of CB HCT.


Chemotaxis/drug effects , Chromatin Assembly and Disassembly/drug effects , Glucocorticoids/pharmacology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Receptors, CXCR4/drug effects , Acetylation/drug effects , Animals , Blotting, Western , Chromatin Immunoprecipitation , E1A-Associated p300 Protein/drug effects , E1A-Associated p300 Protein/metabolism , Fetal Blood/cytology , Flow Cytometry , Histone Code/drug effects , Humans , Mice , Microscopy, Confocal , Nuclear Receptor Coactivator 2/drug effects , Nuclear Receptor Coactivator 2/metabolism , Promoter Regions, Genetic , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Response Elements
19.
Endocr Res ; 42(3): 219-231, 2017 Aug.
Article En | MEDLINE | ID: mdl-28318328

PURPOSE: This study was designed to investigate whether estradiol is related to the expression of the chemokine receptor CXCR4 and its activation in lung adenocarcinoma in vitro, since lung adenocarcinomas from premenopausal women have shown high levels of CXCR4, and this expression has been associated with worse prognosis and poor survival. METHODS: The effect of 17-ß-estradiol (E2) (0.03 nM-10 nM) on CXCR4 expression was analyzed in lung adenocarcinoma cell lines (SK-LU-1, H1435, H23, A549) by immunofluorescence after 24 and 72-h poststimulation. Tamoxifen treatment was applied to corroborate the estrogenic effect. The wound-healing assay was performed to investigate whether E2 treatment increased CXCR4/CXL12 pathway activity. A549 and SK-LU-1 cells were stimulated with E2, CXCL12, and CXCL12 combined with E2. Tamoxifen and AMD3100 were applied to corroborate estrogen and chemokine pathway activation. RESULTS: Estradiol stimulated significantly CXCR4 overexpression in all the cell lines analyzed in a dose- and a time-dependent manner. Tamoxifen treatment inhibited the CXCR4 overexpression observed in estrogen-treated groups, demonstrating that estrogen strongly influences CXCR4 expression in lung adenocarcinoma cells. Cells treated with E2, CXCL12 and E2 combined with CXCL12 exhibited significant cell migration, which was suppressed when tamoxifen and AMD3100 were present. CONCLUSION: Overexpression of CXCR4 induced by estrogen and the activity of CXCL12/CXCR4 pathway could be a new mechanism by which this hormone supports tumor progression and metastasis. These findings may partly explain the worse prognosis observed in premenopausal women and suggest considering the role of estrogen in lung cancer for the design of more specific treatment schemes.


Adenocarcinoma/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Chemokine CXCL12/metabolism , Estradiol/metabolism , Lung Neoplasms/metabolism , Receptors, CXCR4/metabolism , Signal Transduction , Tamoxifen/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma of Lung , Cell Line, Tumor , Female , Humans , Lung Neoplasms/drug therapy , Male , Middle Aged , Receptors, CXCR4/drug effects
20.
Int J Hematol ; 105(4): 423-432, 2017 Apr.
Article En | MEDLINE | ID: mdl-27873175

Combination use of the proteasome inhibitor bortezomib and the immunomodulatory drugs lenalidomide or thalidomide has provided superior outcomes in multiple myeloma over their single use; however, these combinations can produce significant toxicities. Unexpectedly, we found a small but significant increase in the population of immature granulocytes and erythrocytes/megakaryocytes in peripheral blood in 16 of 22 patients (73%) treated with dexamethasone in combination with bortezomib and immunomodulatory drugs (triplet), but not in any of 25 patients treated with either bortezomib or immunomodulatory drugs with dexamethasone (doublet). These immature cells gradually increased to a peak level (mean 2.6% per white blood cells) with triplet therapy, and disappeared immediately after therapy cessation. The numbers of circulating CD34+ cells and colony-forming cells derived from peripheral blood mononuclear cells increased after triplet therapy compared with those in patients treated by either bortezomib or immunomodulatory drugs plus dexamethasone. Furthermore, triplet regimen downregulated the expression of CXCR4, a chemokine receptor essential for bone marrow retention, on CD34+ cells, suggesting an unexpected effect on normal hematopoietic stem/progenitor cells through the reduced interaction with the bone marrow microenvironment. Our observations suggest that combination use should be carefully evaluated to exert synergistic anti-myeloma effects while avoiding unexpected adverse events.


Bortezomib/adverse effects , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/drug effects , Immunologic Factors/adverse effects , Aged , Antigens, CD34/drug effects , Blood Cells/drug effects , Bone Marrow Cells/drug effects , Bortezomib/therapeutic use , Dexamethasone/adverse effects , Dexamethasone/therapeutic use , Drug Therapy, Combination , Female , Humans , Immunologic Factors/therapeutic use , Lenalidomide , Male , Middle Aged , Multiple Myeloma/drug therapy , Receptors, CXCR4/drug effects , Thalidomide/adverse effects , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use
...