Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 156
Filter
1.
Front Aging ; 5: 1414866, 2024.
Article in English | MEDLINE | ID: mdl-38895099

ABSTRACT

Despite effective control of HIV replication by antiretroviral therapy (ART), a significant number of people living with HIV (PLWH) fail to achieve complete immune reconstitution and thus are deemed immune non-responders (INRs). Compared with immune responders (IRs) who have restored their CD4 T cell numbers and functions, CD4 T cells from these INRs exhibit prominent mitochondrial dysfunction and premature aging, which play a major role in increasing the incidence of non-AIDS, non-communicable diseases (NCDs). To date, there are no reliable biomarkers that can be used to typify and manage PLWH, especially INRs with non-AIDS NCDs. Growth differential factor-15 (GDF-15) is a transforming growth factor-ß (TGF-ß) family member known to regulate several biological processes involved in cell aging and stress responses. Since PLWH exhibit premature aging and metabolic dysregulation, here we measured the plasma levels of GDF-15 by ELISA and metabolic proteins by proteomic array and correlated the results with clinical parameters in ART-controlled PLWH (including INRs and IRs) and healthy subjects (HS). We found that GDF-15 levels were significantly elevated in PLWH compared to HS. GDF-15 levels were positively correlated with age and negatively associated with body mass and LDL cholesterol levels in the study subjects. Also, elevated GDF-15 levels were correlated with differential dysregulation of multiple metabolic proteins in PLWH. These results suggest that GDF-15 protein may serve as a biomarker of metabolic dysregulation and aging, and this biomarker will be useful in clinical trials targeting aging and metabolic disorders in ART-treated PLWH.

2.
Ir J Med Sci ; 193(1): 35-43, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37340227

ABSTRACT

BACKGROUND: Doublecortin-like kinase 3 (DCLK3), a member of tubulin superfamily, has been proved to be closely associated with the pathogenesis of numerous human tumors. However, the expression pattern and regulatory mechanisms of DCLK3 in gastric cancer (GC) remain unknown. MATERIALS AND METHODS: DCLK3 expression in GC cells was assessed by RT-qPCR and western blotting. The correlation between DCLK3 levels and the overall survival of GC patients was assessed via TCGA, ACLBI, and Kaplan-Meier plotter databases. Additionally, key proteins (TCF4) involved in the regulation of DCLK3 on GC progression were screened by ACLBI database. Cell proliferation, ferroptotic cell death, and oxidative stress markers were measured by EdU staining, immunofluorescence, ELISA, and western blotting assays. RESULTS: DCLK3 was upregulated in GC, and high DCLK3 expression was significantly associated with poor survival of GC patients. Here, DCLK3 knockdown reduced GC cell proliferation, induced ferroptotic cell death, and exacerbated oxidative stress level. Logistic regression analysis showed that TCF4 was an independent prognostic indicator of GC. Mechanistically, DCLK3 promoted TCF4 expression and subsequently upregulated the expression of TCF4 downstream target genes (c-Myc and Cyclin D1). Furthermore, DCLK3 overexpression enhanced GC cell proliferation, but mitigating ferroptotic cell death and oxidative stress. The regulatory mechanism may involve the upregulation of TCF4, c-Myc, and cyclin D1. CONCLUSIONS: Our research suggests that DCLK3 modulates the levels of iron and reactive oxygen and may involve regulation of TCF4 pathway, thereby promoting the GC cell growth, indicating that DCLK3 may use as a prognostic marker and therapeutic target for GC patients.


Subject(s)
Ferroptosis , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Cyclin D1/genetics , Cyclin D1/metabolism , Cell Line, Tumor , Mitochondria/metabolism , Doublecortin Domain Proteins , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic
3.
J Virol ; 97(11): e0095323, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37877721

ABSTRACT

IMPORTANCE: To our knowledge, this is the first report delineating the activation of the master antioxidant defense during EBV latency. We show that EBV-triggered reactive oxygen species production activates the Keap1-NRF2 pathway in EBV-transformed cells, and LMP1 plays a major role in this event, and the stress-related kinase TBK1 is required for NRF2 activation. Moreover, we show that the Keap1-NRF2 pathway is important for cell proliferation and EBV latency maintenance. Our findings disclose how EBV controls the balance between oxidative stress and antioxidant defense, which greatly improve our understanding of EBV latency and pathogenesis and may be leveraged to opportunities toward the improvement of therapeutic outcomes in EBV-associated diseases.


Subject(s)
Antioxidants , Epstein-Barr Virus Infections , Herpesvirus 4, Human , Latent Infection , Virus Latency , Humans , Antioxidants/metabolism , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/pathogenicity , Herpesvirus 4, Human/physiology , Kelch-Like ECH-Associated Protein 1/metabolism , Latent Infection/metabolism , Latent Infection/virology , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Cell Proliferation
4.
J Transl Med ; 21(1): 682, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37779207

ABSTRACT

BACKGROUND: Recent progress in cancer immunotherapy encourages the expansion of chimeric antigen receptor (CAR) T cell therapy in solid tumors including hepatocellular carcinoma (HCC). Overexpression of MET receptor tyrosine kinase is common in HCC; however, MET inhibitors are effective only when MET is in an active form, making patient stratification difficult. Specific MET-targeting CAR-T cells hold the promise of targeting HCC with MET overexpression regardless of signaling pathway activity. METHODS: MET-specific CARs with CD28ζ or 4-1BBζ as co-stimulation domains were constructed. MET-CAR-T cells derived from healthy subjects (HS) and HCC patients were evaluated for their killing activity and cytokine release against HCC cells with various MET activations in vitro, and for their tumor growth inhibition in orthotopic xenograft models in vivo. RESULTS: MET-CAR.CD28ζ and MET-CAR.4-1BBζ T cells derived from both HS and HCC patients specifically killed MET-positive HCC cells. When stimulated with MET-positive HCC cells in vitro, MET-CAR.CD28ζ T cells demonstrated a higher level of cytokine release and expression of programmed cell death protein 1 (PD-1) than MET-CAR.4-1BBζ T cells. When analyzed in vivo, MET-CAR.CD28ζ T cells more effectively inhibited HCC orthotopic tumor growth in mice when compared to MET-CAR.4-1BBζ T cells. CONCLUSION: We generated and characterized MET-specific CAR-T cells for targeting HCC with MET overexpression regardless of MET activation. Compared with MET-CAR.4-1BBζ, MET-CAR.CD28ζ T cells showed a higher anti-HCC potency but also a higher level of T cell exhaustion. While MET-CAR.CD28ζ is preferred for further development, overcoming the exhaustion of MET-CAR-T cells is necessary to improve their therapeutic efficacy in vivo.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Mice , Animals , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , T-Lymphocytes , Protein-Tyrosine Kinases/metabolism , Cell Line, Tumor , Xenograft Model Antitumor Assays , Immunotherapy, Adoptive , Cytokines/metabolism , Signal Transduction
5.
BMC Nephrol ; 24(1): 245, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37608357

ABSTRACT

BACKGROUND: On December 29, 2021, during the delta wave of the Coronavirus Disease 2019 (COVID-19) pandemic, the stock of premanufactured solutions used for continuous kidney replacement therapy (CKRT) at the University of New Mexico Hospital (UNMH) was nearly exhausted with no resupply anticipated due to supply chain disruptions. Within hours, a backup plan, devised and tested 18 months prior, to locally produce CKRT dialysate was implemented. This report describes the emergency implementation and outcomes of this on-site CKRT dialysate production system. METHODS: This is a single-center retrospective case series and narrative report describing and reporting the outcomes of the implementation of an on-site CKRT dialysate production system. All adults treated with locally produced CKRT dialysate in December 2021 and January 2022 at UNMH were included. CKRT dialysate was produced locally using intermittent hemodialysis machines, hemodialysis concentrate, sterile parenteral nutrition bags, and connectors made of 3-D printed biocompatible rigid material. Outcomes analyzed included dialysate testing for composition and microbiologic contamination, CKRT prescription components, patient mortality, sequential organ failure assessment (SOFA) scores, and catheter-associated bloodstream infections (CLABSIs). RESULTS: Over 13 days, 22 patients were treated with 3,645 L of locally produced dialysate with a mean dose of 20.0 mL/kg/h. Fluid sample testing at 48 h revealed appropriate electrolyte composition and endotoxin levels and bacterial colony counts at or below the lower limit of detection. No CLABSIs occurred within 7 days of exposure to locally produced dialysate. In-hospital mortality was 81.8% and 28-day mortality was 68.2%, though illness severity was high, with a mean SOFA score of 14.5. CONCLUSIONS: Though producing CKRT fluid with IHD machines is not novel, this report represents the first description of the rapid and successful implementation of a backup plan for local CKRT dialysate production at a large academic medical center in the U.S. during the COVID-19 pandemic. Though conclusions are limited by the retrospective design and limited sample size of our analysis, our experience could serve as a guide for other centers navigating similar severe supply constraints in the future.


Subject(s)
COVID-19 , Catheter-Related Infections , Continuous Renal Replacement Therapy , Adult , Humans , Dialysis Solutions , Pandemics , Retrospective Studies
6.
J Med Virol ; 95(7): e28952, 2023 07.
Article in English | MEDLINE | ID: mdl-37455550

ABSTRACT

The presence of hepatitis B virus (HBV) covalently closed circular (ccc) DNA (cccDNA), which serves as a template for viral replication and integration of HBV DNA into the host cell genome, sustains liver pathogenesis and constitutes an intractable barrier to the eradication of chronic HBV infection. The current antiviral therapy for HBV infection, using nucleos(t)ide analogues (NAs), can suppress HBV replication but cannot eliminate integrated HBV DNA and episomal cccDNA. Clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 is a powerful genetic tool that can edit integrated HBV DNA and minichromosomal cccDNA for gene therapy, but its expression and delivery require a viral vector, which poses safety concerns for therapeutic applications in humans. In the present study, we used synthetic guide RNA (gRNA)/Cas9-ribonucleoprotein (RNP) as a nonviral formulation to develop a novel CRISPR/Cas9-mediated gene therapy for eradicating HBV infection. We designed a series of gRNAs targeting multiple specific HBV genes and tested their antiviral efficacy and cytotoxicity in different HBV cellular models. Transfection of stably HBV-infected human hepatoma cell line HepG2.2.15 with HBV-specific gRNA/Cas9 RNPs resulted in a substantial reduction in HBV transcripts. Specifically, gRNA5 and/or gRNA9 RNPs significantly reduced HBV cccDNA, total HBV DNA, pregenomic RNA, and HBV antigen (HBsAg, HBeAg) levels. T7 endonuclease 1 (T7E1) cleavage assay and DNA sequencing confirmed specific HBV gene cleavage and mutations at or around the gRNA target sites. Notably, this gene-editing system did not alter cellular viability or proliferation in the treated cells. Because of their rapid DNA cleavage capability, low off-target effects, low risk of insertional mutagenesis, and readiness for use in clinical application, these results suggest that synthetic gRNA/Cas9 RNP-based gene-editing can be utilized as a promising therapeutic drug for eradicating chronic HBV infection.


Subject(s)
Hepatitis B, Chronic , Hepatitis B , Humans , DNA, Viral/genetics , DNA, Viral/metabolism , CRISPR-Cas Systems , Hepatitis B virus/genetics , Virus Replication , RNA/metabolism , RNA/pharmacology , DNA, Circular/genetics
7.
Viruses ; 15(5)2023 04 26.
Article in English | MEDLINE | ID: mdl-37243148

ABSTRACT

We have previously demonstrated mitochondrial dysfunction in aging CD4 T cells from antiretroviral therapy (ART)-controlled people living with HIV (PLWH). However, the underlying mechanisms by which CD4 T cells develop mitochondrial dysfunction in PLWH remain unclear. In this study, we sought to elucidate the mechanism(s) of CD4 T cell mitochondrial compromise in ART-controlled PLWH. We first assessed the levels of reactive oxygen species (ROS), and we observed significantly increased cellular and mitochondrial ROS levels in CD4 T cells from PLWH compared to healthy subjects (HS). Furthermore, we observed a significant reduction in the levels of proteins responsible for antioxidant defense (superoxide dismutase 1, SOD1) and ROS-mediated DNA damage repair (apurinic/apyrimidinic endonuclease 1, APE1) in CD4 T cells from PLWH. Importantly, CRISPR/Cas9-mediated knockdown of SOD1 or APE1 in CD4 T cells from HS confirmed their roles in maintaining normal mitochondrial respiration via a p53-mediated pathway. Reconstitution of SOD1 or APE1 in CD4 T cells from PLWH successfully rescued mitochondrial function as evidenced by Seahorse analysis. These results indicate that ROS induces mitochondrial dysfunction, leading to premature T cell aging via dysregulation of SOD1 and APE1 during latent HIV infection.


Subject(s)
CD4-Positive T-Lymphocytes , HIV Infections , Humans , Reactive Oxygen Species/metabolism , CD4-Positive T-Lymphocytes/metabolism , HIV Infections/drug therapy , HIV Infections/metabolism , Superoxide Dismutase-1/metabolism , Mitochondria/metabolism
8.
Zhonghua Yi Xue Za Zhi ; 103(6): 425-430, 2023 Feb 14.
Article in Chinese | MEDLINE | ID: mdl-36775266

ABSTRACT

Objective: To investigate the effect of pelvic peritoneum reconstruction on the prognosis of patients underwent laparoscopic low anterior resection of rectal adenocarcinoma. Methods: This retrospective cohort study included 97 patients who underwent laparoscopic low anterior resection of rectal adenocarcinoma in General Surgery Department, the First Affiliated Hospital of Soochow University from January 2017 to June 2021. According to the implementation of pelvic peritoneum reconstruction, the subjects were divided into study group (48 patients underwent pelvic peritoneum reconstruction after laparoscopic resection) and control group (49 patients not underwent pelvic peritoneum reconstruction). The two groups were compared in terms of Wexner score of anal function, anorectal manometry results, normal rate of defecation sensation, pelvic floor anatomical structure and postoperative complications. Five-year survival analysis was performed. Results: Patients in the study group and the control group were (61.25±10.38) years old and (59.47±11.40) years old (P>0.05). The proportions of male patients were 60.4% (29 cases) and 55.1% (27 cases) (P>0.05) in the study group and control group, respectively. At 3, 6 and 12 months after surgery, Wexner scores of anal function in the study group were lower than those in the control group [(14.29±2.07) vs (16.33±2.18), P<0.001; (9.57±2.34) vs (11.26±2.85), P=0.002; (5.41±1.36) vs (7.86±1.95), P<0.001, respectively]. The anal resting pressure and anal systolic pressure of the study group were higher than those of the control group [(56.29±7.31) mmHg vs (52.88±6.65) mmHg, P=0.018; (129.33±17.36) mmHg vs (110.45±15.22) mmHg, P<0.001, respectively] (1 mmHg=0.133 kPa). The rectal sensory volume, rectal maximum tolerance volume, and the length of anal high-pressure area in the study group were greater than those in the control group [(32.15±4.38) vs (29.76±4.29), P=0.008; (209.57±40.27) vs (184.39±37.56), P=0.002; (3.07±0.52) vs (2.80±0.49), P=0.010, respectively]. At 3 and 6 months after surgery, the normal rates of defecation sensation in the study group were 47.9% (23 cases) and 70.8% (34 cases), respectively, higher than those in the control group [26.5% (13 cases) and 51.0% (25 cases)] (P=0.029 and 0.046, respectively). The detection rate of intestinal tube accumulation in the study group was lower than that in the control group [12.5% (6 cases) vs 38.9% (19 cases)] (P=0.003). There was no significant difference in the total incidence of complications (anastomotic leakage, abdominal infection, intestinal obstruction, pendant pneumonia and urinary tract infection) between the two groups [18.8% (9 cases) vs 24.5% (12 cases)] (P=0.493). There was no significant difference in 5-year cumulative survival rate between the study group and the control group (71.6% vs 68.2%, P=0.309). Conclusion: Pelvic peritoneum reconstruction can improve postoperative anal function and reduce intestinal tube accumulation in patients underwent laparoscopic low anterior resection of rectal adenocarcinoma with high safety and feasibility.


Subject(s)
Adenocarcinoma , Laparoscopy , Rectal Neoplasms , Humans , Male , Middle Aged , Aged , Peritoneum/pathology , Retrospective Studies , Rectal Neoplasms/surgery , Prognosis , Laparoscopy/adverse effects , Anal Canal/pathology , Anal Canal/surgery , Pelvic Floor/pathology , Adenocarcinoma/surgery , Adenocarcinoma/pathology , Treatment Outcome
9.
Ultrasonics ; 131: 106961, 2023 May.
Article in English | MEDLINE | ID: mdl-36812819

ABSTRACT

Sonothrombolysis is a technique that utilises ultrasound waves to excite microbubbles surrounding a clot. Clot lysis is achieved through mechanical damage induced by acoustic cavitation and through local clot displacement induced by acoustic radiation force (ARF). Despite the potential of microbubble-mediated sonothrombolysis, the selection of the optimal ultrasound and microbubble parameters remains a challenge. Existing experimental studies are not able to provide a complete picture of how ultrasound and microbubble characteristics influence the outcome of sonothrombolysis. Likewise, computational studies have not been applied in detail in the context of sonothrombolysis. Hence, the effect of interaction between the bubble dynamics and acoustic propagation on the acoustic streaming and clot deformation remains unclear. In the present study, we report for the first time the computational framework that couples the bubble dynamic phenomena with the acoustic propagation in a bubbly medium to simulate microbubble-mediated sonothrombolysis using a forward-viewing transducer. The computational framework was used to investigate the effects of ultrasound properties (pressure and frequency) and microbubble characteristics (radius and concentration) on the outcome of sonothrombolysis. Four major findings were obtained from the simulation results: (i) ultrasound pressure plays the most dominant role over all the other parameters in affecting the bubble dynamics, acoustic attenuation, ARF, acoustic streaming, and clot displacement, (ii) smaller microbubbles could contribute to a more violent oscillation and improve the ARF simultaneously when they are stimulated at higher ultrasound pressure, (iii) higher microbubbles concentration increases the ARF, and (iv) the effect of ultrasound frequency on acoustic attenuation is dependent on the ultrasound pressure. These results may provide fundamental insight that is crucial in bringing sonothrombolysis closer to clinical implementation.


Subject(s)
Computer Simulation , Endovascular Procedures , Mechanical Thrombolysis , Microbubbles , Mechanical Thrombolysis/methods , Ultrasonics , Acoustics
10.
Mol Immunol ; 152: 215-223, 2022 12.
Article in English | MEDLINE | ID: mdl-36379129

ABSTRACT

Identification of immunologic epitopes against SARS-CoV-2 is crucial for the discovery of diagnostic, therapeutic, and preventive targets. In this study, we used a pan-coronavirus peptide microarray to screen for potential B-cell epitopes and validated the results with peptide-based ELISA. Specifically, we identified three linear B-cell epitopes on the SARS-CoV-2 proteome, which were recognized by convalescent plasma from COVID-19 patients. Interestingly, two epitopes (S 809-823 and R1ab 909-923) strongly reacted to convalescent plasma collected at the early phase (< 90 days) of COVID-19 symptom onset, whereas one epitope (M 5-19) reacted to convalescent plasma collected > 90 days after COVID-19 symptom onset. Neutralization assays using antibody depletion with the identified spike (S) peptides revealed that three S epitopes (S 557-571, S 789-803, and S 809-823) elicited neutralizing antibodies in COVID-19 patients. However, the levels of virus-specific antibody targeting S 789-803 only positively correlated with the neutralizing rates at the early phase (<60 days) after disease onset, and the antibody titers diminished quickly with no correlation to the neutralizing activity beyond two months after recovery from COVID-19. Importantly, stimulation of peripheral blood mononuclear cells from COVID-19-recovered patients with these SARS-CoV-2 S peptides resulted in poor virus-specific B cell activation, proliferation, differentiation into memory B cells, and production of immunoglobulin G (IgG) antibodies, despite the B-cells being functionally competent as demonstrated by their response to non-specific stimulation. Taken together, these findings indicate that these newly identified SARS-CoV-2-specific B-cell epitopes can elicit neutralizing antibodies, with titers and/or neutralizing activities declining significantly within 2-3 months in the convalescent plasma of COVID-19 patients.


Subject(s)
COVID-19 , Humans , COVID-19/therapy , SARS-CoV-2 , Epitopes, B-Lymphocyte , Spike Glycoprotein, Coronavirus , Leukocytes, Mononuclear , Antibodies, Viral , Antibodies, Neutralizing , COVID-19 Serotherapy
11.
Front Cell Infect Microbiol ; 12: 1026293, 2022.
Article in English | MEDLINE | ID: mdl-36405960

ABSTRACT

T cells are crucial for controlling viral infections; however, the mechanisms that dampen their responses during viral infections remain incompletely understood. Here, we studied the role and mechanisms of mitochondrial topoisomerase 1 (Top1mt) inhibition in mitochondrial dysfunction and T cell dysregulation using CD4 T cells from patients infected with HCV or HIV and compared it with CD4 T cells from healthy individuals following treatment with Top1 inhibitor - camptothecin (CPT). We found that Top1mt protein levels and enzymatic activity are significantly decreased, along with Top1 cleavage complex (Top1cc) formation, in mitochondria of CD4 T cells from HCV- and HIV-infected patients. Notably, treatment of healthy CD4 T cells with CPT caused similar changes, including inhibition of Top1mt, accumulation of Top1cc in mitochondria, increase in PARP1 cleavage, and decrease in mtDNA copy numbers. These molecular changes resulted in mitochondrial dysfunction, T cell dysregulation, and programmed cell death through multiple signaling pathways, recapitulating the phenotype we detected in CD4 T cells from HCV- and HIV-infected patients. Moreover, treatment of CD4 T cells from HCV or HIV patients with CPT further increased cellular and mitochondrial reactive oxygen species (ROS) production and cell apoptosis, demonstrating a critical role for Top1 in preventing mtDNA damage and cell death. These results provide new insights into the molecular mechanisms underlying immune dysregulation during viral infection and indicate that Top1 inhibition during chronic HCV or HIV infection can induce mtDNA damage and T cell dysfunction. Thus, reconstituting Top1mt protein may restore the mtDNA topology and T cell functions in humans with chronic viral infection.


Subject(s)
HIV Infections , Hepatitis C , Humans , HIV Infections/metabolism , DNA, Mitochondrial/metabolism , DNA Damage , Mitochondria/metabolism
12.
Viruses ; 14(9)2022 08 28.
Article in English | MEDLINE | ID: mdl-36146709

ABSTRACT

The current antiretroviral therapy (ART) for human immunodeficiency virus (HIV) can halt viral replication but cannot eradicate HIV infection because proviral DNA integrated into the host genome remains genetically silent in reservoir cells and is replication-competent upon interruption or cessation of ART. CRISPR/Cas9-based technology is widely used to edit target genes via mutagenesis (i.e., nucleotide insertion/deletion and/or substitution) and thus can inactivate integrated proviral DNA. However, CRISPR/Cas9 delivery systems often require viral vectors, which pose safety concerns for therapeutic applications in humans. In this study, we used synthetic guide RNA (gRNA)/Cas9-ribonucleoprotein (RNP) as a non-viral formulation to develop a novel HIV gene therapy. We designed a series of gRNAs targeting different HIV genes crucial for HIV replication and tested their antiviral efficacy and cellular cytotoxicity in lymphoid and monocytic latent HIV cell lines. Compared with the scramble gRNA control, HIV-gRNA/Cas9 RNP-treated cells exhibited efficient viral suppression with no apparent cytotoxicity, as evidenced by the significant inhibition of latent HIV DNA reactivation and RNA replication. Moreover, HIV-gRNA/Cas9 RNP inhibited p24 antigen expression, suppressed infectious viral particle production, and generated specific DNA cleavages in the targeted HIV genes that are confirmed by DNA sequencing. Because of its rapid DNA cleavage, low off-target effects, low risk of insertional mutagenesis, easy production, and readiness for use in clinical application, this study provides a proof-of-concept that synthetic gRNA/Cas9 RNP drugs can be utilized as a novel therapeutic approach for HIV eradication.


Subject(s)
HIV Infections , HIV-1 , Antiviral Agents , CRISPR-Cas Systems , DNA , HIV-1/genetics , HIV-1/metabolism , Humans , Nucleotides/metabolism , Proviruses/genetics , RNA, Guide, Kinetoplastida/genetics , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Virus Latency
14.
Proteomics Clin Appl ; 16(5): e2200031, 2022 09.
Article in English | MEDLINE | ID: mdl-35929818

ABSTRACT

BACKGROUND: While the majority of COVID-19 patients fully recover from the infection and become asymptomatic, a significant proportion of COVID-19 survivors experience a broad spectrum of symptoms lasting weeks to months post-infection, a phenomenon termed "post-acute sequelae of COVID-19 (PASC)." The aim of this study is to determine whether inflammatory proteins are dysregulated and can serve as potential biomarkers for systemic inflammation in COVID-19 survivors. METHODS: We determined the levels of inflammatory proteins in plasma from 22 coronavirus disease 2019 (COVID-19) long haulers (COV-LH), 22 COVID-19 asymptomatic survivors (COV-AS), and 22 healthy subjects (HS) using an Olink proteomics assay and assessed the results by a beads-based multiplex immunoassay. RESULTS: Compared to HS, we found that COVID-19 survivors still exhibited systemic inflammation, as evidenced by significant changes in the levels of multiple inflammatory proteins in plasma from both COV-LH and COV-AS. CXCL10 was the only protein that significantly upregulated in COV-LH compared with COV-AS and HS. CONCLUSIONS: Our results indicate that several inflammatory proteins remain aberrantly dysregulated in COVID-19 survivors and CXCL10 might serve as a potential biomarker to typify COV-LH. Further characterization of these signature inflammatory molecules might improve the understanding of the long-term impacts of COVID-19 and provide new targets for the diagnosis and treatment of COVID-19 survivors with PASC.


Subject(s)
COVID-19 , Biomarkers , COVID-19/complications , Humans , Inflammation , SARS-CoV-2 , Survivors
15.
J Cell Sci ; 135(13)2022 07 01.
Article in English | MEDLINE | ID: mdl-35660868

ABSTRACT

We investigated the role of telomerase and telomere repeat-binding factor 2 (TRF2 or TERF2) in T-cell dysfunction in chronic viral infection. We found that the expression and activity of telomerase in CD4+ T (CD4T) cells from patients with hepatitis C virus (HCV) infections or people living with HIV (PLWH) were intact, but TRF2 expression was significantly inhibited at the post-transcriptional level, suggesting that TRF2 inhibition is responsible for the CD4T cell dysfunction observed during chronic viral infection. Silencing TRF2 expression in CD4T cells derived from healthy subjects induced telomeric DNA damage and CD4T cell dysfunction without affecting telomerase activity or translocation - similar to what we observed in CD4T cells from HCV patients and PLWH. These findings indicate that premature T-cell aging and dysfunction during chronic HCV or HIV infection are primarily caused by chronic immune stimulation and T-cell overactivation and/or proliferation that induce telomeric DNA damage due to TRF2 inhibition, rather than telomerase disruption. This study suggests that restoring TRF2 presents a novel approach to prevent telomeric DNA damage and premature T-cell aging, thus rejuvenating T-cell functions during chronic viral infection.


Subject(s)
CD4-Positive T-Lymphocytes , HIV Infections , Telomerase , Telomeric Repeat Binding Protein 2 , CD4-Positive T-Lymphocytes/immunology , DNA Damage , HIV Infections/genetics , HIV Infections/immunology , Hepacivirus , Hepatitis C, Chronic/genetics , Hepatitis C, Chronic/immunology , Humans , Telomerase/genetics , Telomerase/metabolism , Telomere , Telomeric Repeat Binding Protein 2/antagonists & inhibitors , Telomeric Repeat Binding Protein 2/genetics , Telomeric Repeat Binding Protein 2/metabolism
17.
Front Immunol ; 13: 837524, 2022.
Article in English | MEDLINE | ID: mdl-35251030

ABSTRACT

Effectively treating infectious diseases often requires a multi-step approach to target different components involved in disease pathogenesis. Similarly, the COVID-19 pandemic has become a global health crisis that requires a comprehensive understanding of Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2) infection to develop effective therapeutics. One potential strategy to instill greater immune protection against COVID-19 is boosting the innate immune system. This boosting, termed trained immunity, employs immune system modulators to train innate immune cells to produce an enhanced, non-specific immune response upon reactivation following exposure to pathogens, a process that has been studied in the context of in vitro and in vivo clinical studies prior to the COVID-19 pandemic. Evaluation of the underlying pathways that are essential to inducing protective trained immunity will provide insight into identifying potential therapeutic targets that may alleviate the COVID-19 crisis. Here we review multiple immune training agents, including Bacillus Calmette-Guérin (BCG), ß-glucan, and lipopolysaccharide (LPS), and the two most popular cell types involved in trained immunity, monocytes and natural killer (NK) cells, and compare the signaling pathways involved in innate immunity. Additionally, we discuss COVID-19 trained immunity clinical trials, emphasizing the potential of trained immunity to fight SARS-CoV-2 infection. Understanding the mechanisms by which training agents activate innate immune cells to reprogram immune responses may prove beneficial in developing preventive and therapeutic targets against COVID-19.


Subject(s)
COVID-19/immunology , Immunity, Innate/immunology , Humans , Killer Cells, Natural/immunology , Monocytes/immunology , SARS-CoV-2/immunology
18.
Front Immunol ; 13: 823660, 2022.
Article in English | MEDLINE | ID: mdl-35185915

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) prolong sepsis by promoting immunosuppression. We reported that sepsis MDSC development requires long non-coding RNA Hotairm1 interactions with S100A9. Using a mouse model that simulates the immunobiology of sepsis, we find that histone demethylase KDM6A promotes Hotairm1 transcription by demethylating transcription repression H3K27me3 histone mark. We show that chemical targeting of KDM6A by GSK-J4 represses Hotairm1 transcription, which coincides with decreases in transcription activation H3K4me3 histone mark and transcription factor PU.1 binding to the Hotairm1 promoter. We further show that immunosuppressive IL-10 cytokine promotes KDM6A binding at the Hotairm1 promoter. IL-10 knockdown repletes H3K27me3 and reduces Hotairm1 transcription. GSK-J4 treatment also relocalizes nuclear S100A9 protein to the cytosol. To support translation to human sepsis, we demonstrate that inhibiting H3K27me3 demethylation by KDM6A ex vivo in MDSCs from patients with protracted sepsis decreases Hotairm1 transcription. These findings suggest that epigenetic targeting of MDSCs in human sepsis might resolve post-sepsis immunosuppression and improve sepsis survival.


Subject(s)
Histone Demethylases/metabolism , MicroRNAs/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Sepsis/metabolism , Sepsis/pathology , Animals , Benzazepines/pharmacology , Calgranulin B/metabolism , Histone Code , Histones/genetics , Histones/metabolism , Humans , Immunosuppression Therapy , Interleukin-10/genetics , Interleukin-10/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Pyrimidines/pharmacology
19.
J Innate Immun ; 14(2): 112-123, 2022.
Article in English | MEDLINE | ID: mdl-34289476

ABSTRACT

Sepsis-induced myeloid-derived suppressor cells (MDSCs) increase mortality risk. We previously identified that long non-coding RNA Hotairm1 supports myeloid precursor shifts to Gr1+CD11b+ MDSCs during mouse sepsis. A major unanswered question is what molecular processes control Hotairm1 expression. In this study, we found by a genetic deletion that a specific PU.1-binding site is indispensable in controlling Hotairm1 transcription. We then identified H3K4me3 and H3K27me3 at the PU.1 site on the Hotairm1 promoter. Controlling an epigenetic switch of Hotairm1 transcription by PU.1 was histone KDM6A demethylase for H3K27me3 that derepressed its transcription with possible contributions from Ezh2 methyltransferase for H3K27me3. KDM6A knockdown in MDSCs increased H3K27me3, decreased H3K4me3, and inhibited Hotairm1 transcription activation by PU.1. These results enlighten clinical translation research of PU.1 epigenetic regulation as a potential sepsis immune-checkpoint treatment site.


Subject(s)
MicroRNAs , Myeloid-Derived Suppressor Cells , Sepsis , Animals , Epigenesis, Genetic , Histone Demethylases/genetics , Histone Demethylases/metabolism , Lysine/genetics , Lysine/metabolism , Mice , MicroRNAs/genetics , Sepsis/genetics , Sepsis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...