Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
FASEB J ; 35(12): e22044, 2021 12.
Article in English | MEDLINE | ID: mdl-34818449

ABSTRACT

Pulmonary arterial hypertension (PAH) is a common and fatal complication of systemic lupus erythematosus (SLE). Whether the BMP receptor deficiency found in the genetic form of PAH is also involved in SLE-PAH patients remains to be identified. In this study, we employed patient-derived samples from SLE-associated PAH (SLE-PAH) and established comparable mouse models to clarify the role of BMP signaling in the pathobiology of SLE-PAH. Firstly, serum levels of LPS and autoantibodies (auto-Abs) directed at BMP receptors were significantly increased in patients with SLE-PAH compared with control subjects, measured by ELISA. Mass cytometry was applied to compare peripheral blood leukocyte phenotype in patients prior to and after treatment with steroids, which demonstrated inflammatory cells alteration in SLE-PAH. Furthermore, BMPR2 signaling and pyroptotic factors were examined in human pulmonary arterial endothelial cells (PAECs) in response to LPS stimulation. Interleukin-8 (IL-8) and E-selectin (SELE) expressions were up-regulated in autologous BMPR2+/R899X endothelial cells and siBMPR2-interfered PAECs. A SLE-PH model was established in mice induced with pristane and hypoxia. Moreover, the combination of endothelial specific BMPR2 knockout in SLE mice exacerbated pulmonary hypertension. Pyroptotic factors including gasdermin D (GSDMD) were elevated in the lungs of SLE-PH mice, and the pyroptotic effects of serum samples isolated from SLE-PAH patients on PAECs were analyzed. BMPR2 signaling upregulator (BUR1) showed anti-pyroptotic effects in SLE-PH mice and PAECs. Our results implied that deficiencies of BMPR2 signaling and proinflammatory factors together contribute to the development of PAH in SLE.


Subject(s)
Autoantibodies/immunology , Bone Morphogenetic Protein Receptors, Type II/deficiency , Endothelial Cells/immunology , Lipopolysaccharides/toxicity , Lupus Erythematosus, Systemic/pathology , Pulmonary Arterial Hypertension/pathology , Pyroptosis , Activin Receptors, Type II/immunology , Adult , Animals , Autoantibodies/blood , Bone Morphogenetic Protein Receptors, Type I/immunology , Bone Morphogenetic Protein Receptors, Type II/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Humans , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/metabolism , Male , Mice , Mice, Inbred BALB C , Pulmonary Arterial Hypertension/etiology , Pulmonary Arterial Hypertension/metabolism , Vascular Remodeling
2.
J Clin Invest ; 131(19)2021 10 01.
Article in English | MEDLINE | ID: mdl-34375315

ABSTRACT

BACKGROUNDChimeric antigen receptor (CAR) T cells have emerged as an approach to treat malignant tumors. This strategy has also been proposed for the treatment of HIV-1 infection. We have developed a broadly neutralizing antibody-derived (bNAb-derived) CAR T cell therapy that can exert specific cytotoxic activity against HIV-1-infected cells.METHODSWe conducted an open-label trial of the safety, side-effect profile, pharmacokinetic properties, and antiviral activity of bNAb-derived CAR T cell therapy in individuals infected with HIV-1 who were undergoing analytical interruption of antiretroviral therapy (ART).RESULTSA total of 14 participants completed only a single administration of bNAb-derived CAR T cells. CAR T cell therapy administration was safe and well tolerated. Six participants discontinued ART, and viremia rebound occurred in all of them, with a 5.3-week median time. Notably, the cell-associated viral RNA and intact proviruses decreased significantly after CAR T cell treatment. Analyses of HIV-1 variants before or after CAR T cell administration suggested that CAR T cells exerted pressure on rebound viruses, resulting in a selection of viruses with less diversity and mutations against CAR T cell-mediated cytotoxicity.CONCLUSIONNo safety concerns were identified with adoptive transfer of bNAb-derived CAR T cells. They reduced viral reservoir. All the rebounds were due to preexisting or emergence of viral escape mutations.TRIAL REGISTRATIONClinicalTrials.gov (NCT03240328).FUNDINGMinistry of Science and Technology of China, National Natural Science Foundation of China, and Department of Science and Technology of Guangdong Province.


Subject(s)
Broadly Neutralizing Antibodies/immunology , HIV Infections/therapy , HIV-1/immunology , Immunotherapy, Adoptive , Receptors, Chimeric Antigen/immunology , Viral Load , Adult , CD4-Positive T-Lymphocytes/immunology , HEK293 Cells , HIV Infections/virology , Humans , Male , Middle Aged
3.
ACS Appl Mater Interfaces ; 13(33): 39621-39632, 2021 Aug 25.
Article in English | MEDLINE | ID: mdl-34383462

ABSTRACT

To achieve high sensitivity under low-temperature operation is currently a challenge for metal oxide semiconductor gas sensors. In this work, a unique NiO-functionalized macroporous In2O3 thin film is designed by atomic layer deposition (ALD), which demonstrates great potential in electronic sensors for detecting NO2 at low temperature. This strategy allows for efficient engineering of the oxygen vacancy concentration and the formation of p-n heterojunctions in the hybrid In2O3/NiO thin films, which has been found to greatly impact the surface chemical and electrical properties of the sensing films. The sensor based on the optimized In2O3/NiO films exhibits a very high response of 532.2 to 10 ppm NO2, which is 26 times higher than that of the In2O3, at a relatively low operating temperature of 145 °C. In addition, an ultralow detection limit of ca. 6.9 ppb has been obtained, which surpasses most reports based on metal oxide sensors. Mechanistic investigations disclose that the improved sensor properties are resultant from the paramount surface active sites and high carrier concentration enabled by the oxygen vacancies, while excessive NiO ALD leads to a decreased sensor response due to the formed p-n heterojunctions.

4.
J Virol ; 94(10)2020 05 04.
Article in English | MEDLINE | ID: mdl-32161174

ABSTRACT

Although substantial progress has been made in depicting the molecular pathogenesis of human immunodeficiency virus type 1 (HIV-1) infection, the comprehensive mechanism of HIV-1 latency and the most promising therapeutic strategies to effectively reactivate the HIV-1 latent reservoir to achieve a functional cure for AIDS remain to be systematically illuminated. Here, we demonstrated that piwi (P element-induced Wimpy)-like RNA-mediated gene silencing 4 (PIWIL4) played an important role in suppressing HIV-1 transcription and contributed to the latency state in HIV-1-infected cells through its recruitment of various suppressive factors, including heterochromatin protein 1α/ß/γ, SETDB1, and HDAC4. The knockdown of PIWIL4 enhanced HIV-1 transcription and reversed HIV-1 latency in both HIV-1 latently infected Jurkat T cells and primary CD4+ T lymphocytes and resting CD4+ T lymphocytes from HIV-1-infected individuals on suppressive combined antiretroviral therapy (cART). Furthermore, in the absence of PIWIL4, HIV-1 latently infected Jurkat T cells were more sensitive to reactivation with vorinostat (suberoylanilide hydroxamic acid, or SAHA), JQ1, or prostratin. These findings indicated that PIWIL4 promotes HIV-1 latency by imposing repressive marks at the HIV-1 5' long terminal repeat. Thus, the manipulation of PIWIL4 could be a novel strategy for developing promising latency-reversing agents (LRAs).IMPORTANCE HIV-1 latency is systematically modulated by host factors and viral proteins. During this process, the suppression of HIV-1 transcription plays an essential role in promoting HIV-1 latency. In this study, we found that PIWIL4 repressed HIV-1 promoter activity and maintained HIV-1 latency. In particular, we report that PIWIL4 can regulate gene expression through its association with the suppressive activity of HDAC4. Therefore, we have identified a new function for PIWIL4: it is not only a suppressor of endogenous retrotransposons but also plays an important role in inhibiting transcription and leading to latent infection of HIV-1, a well-known exogenous retrovirus. Our results also indicate a novel therapeutic target to reactivate the HIV-1 latent reservoir.


Subject(s)
Argonaute Proteins/metabolism , Argonaute Proteins/pharmacology , Epigenesis, Genetic , Gene Expression Regulation, Viral/drug effects , HIV-1/physiology , Virus Latency/drug effects , Anti-Retroviral Agents/therapeutic use , Argonaute Proteins/genetics , CD4-Positive T-Lymphocytes/virology , HEK293 Cells , HIV Infections/virology , HIV-1/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Jurkat Cells , RNA-Binding Proteins , Repressor Proteins/genetics , Repressor Proteins/metabolism , Viral Proteins/metabolism , Virus Latency/genetics , Virus Replication/drug effects
5.
Front Immunol ; 10: 2151, 2019.
Article in English | MEDLINE | ID: mdl-31572371

ABSTRACT

Current combined antiretroviral therapy (cART) mainly targets 3 of the 15 HIV proteins leaving many potential viral vulnerabilities unexploited. To purge the HIV-1 latent reservoir, various strategies including "shock and kill" have been developed. A key question is how to restore impaired immune surveillance. HIV-1 protein Nef has long been known to mediate the downregulation of cell-surface MHC-I and assist HIV-1 to evade the immune system. Through high throughput screening of Food and Drug Administration (FDA) approved drugs, we identified lovastatin, a statin drug, to significantly antagonize Nef to downregulate MHC-I, CD4, and SERINC5, and inhibit the intrinsic infectivity of virions. In addition, lovastatin boosted autologous CTLs to eradicate the infected cells and effectively inhibit the subsequent viral rebound in CD4+ T-lymphocytes isolated from HIV-1-infected individuals receiving suppressive cART. Furthermore, we found that lovastatin inhibits Nef-induced MHC-I downregulation by directly binding with Nef and disrupting the Nef-AP-1 complex. These results demonstrate that lovastatin is a promising agent for counteracting Nef-mediated downregulation of MHC-I, CD4, and SERINC5. Lovastatin could potentially be used in the clinic to enhance anti-HIV-1 immune surveillance.


Subject(s)
HIV Infections/immunology , Histocompatibility Antigens Class I/immunology , Lovastatin/pharmacology , CD4 Antigens/immunology , Cells, Cultured , Coculture Techniques , Down-Regulation , HIV-1 , Humans , Membrane Proteins/immunology , T-Lymphocytes/immunology , Transcription Factor AP-1/immunology , nef Gene Products, Human Immunodeficiency Virus/immunology
6.
Nat Commun ; 10(1): 4109, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31511513

ABSTRACT

The inhibitory receptors PD-1, Tim-3, and Lag-3 are highly expressed on tumor-infiltrating lymphocytes and compromise their antitumor activity. For efficient cancer immunotherapy, it is important to prevent chimeric antigen receptor T (CAR-T)-cell exhaustion. Here we downregulate these three checkpoint receptors simultaneously on CAR-T cells and that show the resulting PTL-CAR-T cells undergo epigenetic modifications and better control tumor growth. Furthermore, we unexpectedly find increased tumor infiltration by PTL-CAR-T cells and their clustering between the living and necrotic tumor tissue. Mechanistically, PTL-CAR-T cells upregulate CD56 (NCAM), which is essential for their effector function. The homophilic interaction between intercellular CD56 molecules correlates with enhanced infiltration of CAR-T cells, increased secretion of interferon-γ, and the prolonged survival of CAR-T cells. Ectopically expressed CD56 promotes CAR-T cell survival and antitumor response. Our findings demonstrate that genetic blockade of three checkpoint inhibitory receptors and the resulting high expression of CD56 on CAR-T cells enhances the inhibition of tumor growth.


Subject(s)
CD56 Antigen/metabolism , Neoplasms/immunology , Neoplasms/therapy , Protein Engineering , Receptors, Chimeric Antigen/metabolism , Animals , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , Female , HEK293 Cells , Humans , Immunotherapy, Adoptive , Mice , Neoplasms/drug therapy , Receptor, ErbB-2/metabolism
7.
Retrovirology ; 15(1): 20, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29426337

ABSTRACT

Retroviruses can cause severe diseases such as cancer and acquired immunodeficiency syndrome. A unique feature in the life cycle of retroviruses is that their RNA genome is reverse transcribed into double-stranded DNA, which then integrates into the host genome to exploit the host machinery for their benefits. The metazoan genome encodes numerous non-coding RNAs (ncRNA), which act as key regulators in essential cellular processes such as antiviral response. The development of next-generation sequencing technology has greatly accelerated the detection of ncRNAs from viruses and their hosts. ncRNAs have been shown to play important roles in the retroviral life cycle and virus-host interactions. Here, we review recent advances in ncRNA studies with special focus on those have changed our understanding of retroviruses or provided novel strategies to treat retrovirus-related diseases. Many ncRNAs such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are involved in the late phase of the retroviral life cycle. However, their roles in the early phase of viral replication merit further investigations.


Subject(s)
RNA, Untranslated/physiology , Retroviridae Infections/virology , Retroviridae/genetics , Retroviridae/physiology , Gene Expression Regulation, Viral , Host-Pathogen Interactions , Humans , MicroRNAs , RNA Interference , RNA, Long Noncoding , Virus Latency
SELECTION OF CITATIONS
SEARCH DETAIL
...