Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2807: 93-110, 2024.
Article in English | MEDLINE | ID: mdl-38743223

ABSTRACT

Correlative light-electron microscopy (CLEM) has evolved in the last decades, especially after significant developments in sample preparation, imaging acquisition, software, spatial resolution, and equipment, including confocal, live-cell, super-resolution, and electron microscopy (scanning, transmission, focused ion beam, and cryo-electron microscopy). However, the recent evolution of different laser-related techniques, such as mass spectrometry imaging (MSI) and laser capture microdissection, could further expand spatial imaging capabilities into high-resolution OMIC approaches such as proteomic, lipidomics, small molecule, and drug discovery. Here, we will describe a protocol to integrate the detection of rare viral reservoirs with imaging mass spectrometry.


Subject(s)
HIV Infections , Humans , HIV Infections/virology , HIV-1/physiology , Mass Spectrometry/methods , Microscopy, Electron/methods , Molecular Imaging/methods , Disease Reservoirs/virology
2.
MicroPubl Biol ; 20242024.
Article in English | MEDLINE | ID: mdl-38404916

ABSTRACT

UNC45B is a multidomain molecular chaperone that is essential for the proper folding and function of myosin. It has previously been demonstrated that the UCS domain is responsible for the chaperoning function of UNC45B and that removing its client-binding loop leads to a significant change in its solution conformation and a reduced chaperoning function. Here, we report the direct quantification of affinities of myosin binding to wild type and mutant UNC45B using surface plasmon resonance (SPR) spectroscopy. We found that deletion of the client-binding loop in UNC45B resulted in a dramatic decrease in myosin affinity.

3.
Cell Rep ; 42(11): 113285, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37910505

ABSTRACT

Deciphering the mechanisms underlying viral persistence is critical to achieving a cure for human immunodeficiency virus (HIV) infection. Here, we implement a systems approach to discover molecular signatures of HIV latently infected CD4+ T cells, identifying the immunosuppressive, adenosine-producing ectonucleotidase CD73 as a key surface marker of latent cells. Hypoxic conditioning, reflecting the lymphoid tissue microenvironment, increases the frequency of CD73+ CD4+ T cells and promotes HIV latency. Transcriptomic profiles of CD73+ CD4+ T cells favor viral quiescence, immune evasion, and cell survival. CD73+ CD4+ T cells are capable of harboring a functional HIV reservoir and reinitiating productive infection ex vivo. CD73 or adenosine receptor blockade facilitates latent HIV reactivation in vitro, mechanistically linking adenosine signaling to viral quiescence. Finally, tissue imaging of lymph nodes from HIV-infected individuals on antiretroviral therapy reveals spatial association between CD73 expression and HIV persistence in vivo. Our findings warrant development of HIV-cure strategies targeting the hypoxia-CD73-adenosine axis.


Subject(s)
HIV Infections , HIV-1 , Humans , Adenosine/metabolism , CD4-Positive T-Lymphocytes , Virus Activation , Virus Latency/physiology , Virus Replication/physiology
4.
NeuroImmune Pharm Ther ; 2(2): 169-186, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37476291

ABSTRACT

Tunneling nanotubes (TNTs), also called cytonemes or tumor microtubes, correspond to cellular processes that enable long-range communication. TNTs are plasma membrane extensions that form tubular processes that connect the cytoplasm of two or more cells. TNTs are mostly expressed during the early stages of development and poorly expressed in adulthood. However, in disease conditions such as stroke, cancer, and viral infections such as HIV, TNTs proliferate, but their role is poorly understood. TNTs function has been associated with signaling coordination, organelle sharing, and the transfer of infectious agents such as HIV. Here, we describe the critical role and function of TNTs during HIV infection and reactivation, as well as the use of TNTs for cure strategies.

5.
Cells ; 11(15)2022 08 02.
Article in English | MEDLINE | ID: mdl-35954221

ABSTRACT

The major barrier to cure HIV infection is the early generation and extended survival of HIV reservoirs in the circulation and tissues. Currently, the techniques used to detect and quantify HIV reservoirs are mostly based on blood-based assays; however, it has become evident that viral reservoirs remain in tissues. Our study describes a novel multi-component imaging method (HIV DNA, mRNA, and viral proteins in the same assay) to identify, quantify, and characterize viral reservoirs in tissues and blood products obtained from HIV-infected individuals even when systemic replication is undetectable. In the human brains of HIV-infected individuals under ART, we identified that microglia/macrophages and a small population of astrocytes are the main cells with integrated HIV DNA. Only half of the cells with integrated HIV DNA expressed viral mRNA, and one-third expressed viral proteins. Surprisingly, we identified residual HIV-p24, gp120, nef, vpr, and tat protein expression and accumulation in uninfected cells around HIV-infected cells suggesting local synthesis, secretion, and bystander uptake. In conclusion, our data show that ART reduces the size of the brain's HIV reservoirs; however, local/chronic viral protein secretion still occurs, indicating that the brain is still a major anatomical target to cure HIV infection.


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , Brain , DNA , Humans , RNA, Messenger , Viral Proteins , Virus Latency
6.
Cell Mol Life Sci ; 79(7): 365, 2022 Jun 16.
Article in English | MEDLINE | ID: mdl-35708858

ABSTRACT

SARS-CoV-2, although not being a circulatory virus, spread from the respiratory tract resulting in multiorgan failures and thrombotic complications, the hallmarks of fatal COVID-19. A convergent contributor could be platelets that beyond hemostatic functions can carry infectious viruses. Here, we profiled 52 patients with severe COVID-19 and demonstrated that circulating platelets of 19 out 20 non-survivor patients contain SARS-CoV-2 in robust correlation with fatal outcome. Platelets containing SARS-CoV-2 might originate from bone marrow and lung megakaryocytes (MKs), the platelet precursors, which were found infected by SARS-CoV-2 in COVID-19 autopsies. Accordingly, MKs undergoing shortened differentiation and expressing anti-viral IFITM1 and IFITM3 RNA as a sign of viral sensing were enriched in the circulation of deadly COVID-19. Infected MKs reach the lung concomitant with a specific MK-related cytokine storm rich in VEGF, PDGF and inflammatory molecules, anticipating fatal outcome. Lung macrophages capture SARS-CoV-2-containing platelets in vivo. The virus contained by platelets is infectious as capture of platelets carrying SARS-CoV-2 propagates infection to macrophages in vitro, in a process blocked by an anti-GPIIbIIIa drug. Altogether, platelets containing infectious SARS-CoV-2  alter COVID-19 pathogenesis and provide a powerful fatality marker. Clinical targeting of platelets might prevent viral spread, thrombus formation and exacerbated inflammation at once and increase survival in COVID-19.


Subject(s)
COVID-19 , Thrombosis , Blood Platelets , Humans , Lung , Megakaryocytes , Membrane Proteins , RNA-Binding Proteins , SARS-CoV-2
7.
iScience ; 24(12): 103478, 2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34841222

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) rapidly rampaged worldwide, causing a pandemic of coronavirus disease (COVID -19), but the biology of SARS-CoV-2 remains under investigation. We demonstrate that both SARS-CoV-2 spike protein and human coronavirus 229E (hCoV-229E) or its purified S protein, one of the main viruses responsible for the common cold, induce the transient opening of Pannexin-1 (Panx-1) channels in human lung epithelial cells. However, the Panx-1 channel opening induced by SARS-CoV-2 is greater and more prolonged than hCoV-229E/S protein, resulting in an enhanced ATP, PGE2, and IL-1ß release. Analysis of lung lavages and tissues indicate that Panx-1 mRNA expression is associated with increased ATP, PGE2, and IL-1ß levels. Panx-1 channel opening induced by SARS-CoV-2 spike protein is angiotensin-converting enzyme 2 (ACE-2), endocytosis, and furin dependent. Overall, we demonstrated that Panx-1 channel is a critical contributor to SARS-CoV-2 infection and should be considered as an alternative therapy.

8.
Front Immunol ; 12: 735922, 2021.
Article in English | MEDLINE | ID: mdl-34671353

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a major public health issue. COVID-19 is considered an airway/multi-systemic disease, and demise has been associated with an uncontrolled immune response and a cytokine storm in response to the virus. However, the lung pathology, immune response, and tissue damage associated with COVID-19 demise are poorly described and understood due to safety concerns. Using post-mortem lung tissues from uninfected and COVID-19 deadly cases as well as an unbiased combined analysis of histology, multi-viral and host markers staining, correlative microscopy, confocal, and image analysis, we identified three distinct phenotypes of COVID-19-induced lung damage. First, a COVID-19-induced hemorrhage characterized by minimal immune infiltration and large thrombus; Second, a COVID-19-induced immune infiltration with excessive immune cell infiltration but no hemorrhagic events. The third phenotype correspond to the combination of the two previous ones. We observed the loss of alveolar wall integrity, detachment of lung tissue pieces, fibroblast proliferation, and extensive fibrosis in all three phenotypes. Although lung tissues studied were from lethal COVID-19, a strong immune response was observed in all cases analyzed with significant B cell and poor T cell infiltrations, suggesting an exhausted or compromised immune cellular response in these patients. Overall, our data show that SARS-CoV-2-induced lung damage is highly heterogeneous. These individual differences need to be considered to understand the acute and long-term COVID-19 consequences.


Subject(s)
COVID-19/mortality , COVID-19/pathology , Lung Injury/pathology , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/pathology , Aged , Aged, 80 and over , Autopsy , CD8-Positive T-Lymphocytes/immunology , Cytokine Release Syndrome/mortality , Cytokine Release Syndrome/pathology , Epithelial Cells/pathology , Female , Hemorrhage/pathology , Humans , Inflammation/pathology , Lung/pathology , Lung Injury/virology , Lymphopenia/pathology , Macrophage Activation/immunology , Macrophages/immunology , Male , Middle Aged , Myocytes, Smooth Muscle/pathology , Neutrophils/immunology , SARS-CoV-2 , Thrombosis/pathology
9.
Purinergic Signal ; 17(4): 563-576, 2021 12.
Article in English | MEDLINE | ID: mdl-34542793

ABSTRACT

Only recently, the role of large ionic channels such as Pannexin-1 channels and Connexin hemichannels has been implicated in several physiological and pathological conditions, including HIV infection and associated comorbidities. These channels are in a closed stage in healthy conditions, but in pathological conditions including HIV, Pannexin-1 channels and Connexin hemichannels become open. Our data demonstrate that acute and chronic HIV infection induces channel opening (Pannexin and Connexin channels), ATP release into the extracellular space, and subsequent activation of purinergic receptors in immune and non-immune cells. We demonstrated that Pannexin and Connexin channels contribute to HIV infection and replication, the long-term survival of viral reservoirs, and comorbidities such as NeuroHIV. Here, we discuss the available data to support the participation of these channels in the HIV life cycle and the potential therapeutic approach to prevent HIV-associated comorbidities.


Subject(s)
Connexins/metabolism , HIV Infections/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Purinergic/metabolism , Animals , Humans
10.
Prog Neurobiol ; 206: 102157, 2021 11.
Article in English | MEDLINE | ID: mdl-34455020

ABSTRACT

HIV-associated neurological dysfunction is observed in more than half of the HIV-infected population, even in the current antiretroviral era. The mechanisms by which HIV mediates CNS dysfunction are not well understood but have been associated with the presence of long-lasting HIV reservoirs. In the CNS, macrophage/microglia and a small population of astrocytes harbor the virus. However, the low number of HIV-infected cells does not correlate with the high degree of damage, suggesting that mechanisms of damage amplification may be involved. Here, we demonstrate that the survival mechanism of HIV-infected cells and the apoptosis of surrounding uninfected cells is regulated by inter-organelle interactions among the mitochondria/Golgi/endoplasmic reticulum system and the associated signaling mediated by IP3 and calcium. We identified that latently HIV-infected astrocytes had elevated intracellular levels of IP3, a master regulator second messenger, which diffuses via gap junctions into neighboring uninfected astrocytes resulting in their apoptosis. In addition, using laser capture microdissection, we confirmed that bystander apoptosis of uninfected astrocytes and the survival of HIV-infected astrocytes were dependent on mitochondrial function, intracellular calcium, and IP3 signaling. Blocking gap junction channels did not prevent an increase in IP3 or inter-organelle dysfunction in HIV-infected cells but reduced the amplification of apoptosis into uninfected neighboring cells. Our data provide a mechanistic explanation for bystander damage induced by surviving infected cells that serve as viral reservoirs and provide potential targets for interventions to reduce the devastating consequences of HIV within the brain.


Subject(s)
HIV Infections , Astrocytes/metabolism , Calcium/metabolism , HIV Infections/complications , HIV Infections/metabolism , Humans , Inositol Phosphates/metabolism , Mitochondria
11.
Sci Rep ; 11(1): 14556, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34267246

ABSTRACT

Cell-to-cell communication is essential for the development and proper function of multicellular systems. We and others demonstrated that tunneling nanotubes (TNT) proliferate in several pathological conditions such as HIV, cancer, and neurodegenerative diseases. However, the nature, function, and contribution of TNT to cancer pathogenesis are poorly understood. Our analyses demonstrate that TNT structures are induced between glioblastoma (GBM) cells and surrounding non-tumor astrocytes to transfer tumor-derived mitochondria. The mitochondrial transfer mediated by TNT resulted in the adaptation of non-tumor astrocytes to tumor-like metabolism and hypoxia conditions. In conclusion, TNT are an efficient cell-to-cell communication system used by cancer cells to adapt the microenvironment to the invasive nature of the tumor.


Subject(s)
Astrocytes/pathology , Glioblastoma/pathology , Mitochondria/pathology , Astrocytes/metabolism , Cell Communication , Cell Hypoxia , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , DNA, Mitochondrial , Humans , Laser Capture Microdissection , Microscopy, Electron, Transmission , Mitochondria/genetics , Oxidative Stress , Tumor Microenvironment
12.
J Neurochem ; 158(2): 500-521, 2021 07.
Article in English | MEDLINE | ID: mdl-33899944

ABSTRACT

In healthy conditions, pannexin-1 (Panx-1) channels are in a close state, but in several pathological conditions, including human immunodeficiency virus-1 (HIV) and NeuroHIV, the channel becomes open. However, the mechanism or contribution of Panx-1 channels to the HIV pathogenesis and NeuroHIV is unknown. To determine the contribution of Panx-1 channels to the pathogenesis of NeuroHIV, we used a well-established model of simian immunodeficiency virus (SIV) infection in macaques (Macaca mulatta) in the presence of and absence of a Panx-1 blocker to later examine the synaptic/axonal compromise induced for the virus. Using Golgi's staining, we demonstrated that SIV infection compromised synaptic and axonal structures, especially in the white matter. Blocking Panx-1 channels after SIV infection prevented the synaptic and axonal compromise induced by the virus, especially by maintaining the more complex synapses. Our data demonstrated that targeting Panx-1 channels can prevent and maybe revert brain synaptic compromise induced by SIV infection.


Subject(s)
Connexins/metabolism , HIV Infections/metabolism , HIV-1 , Nerve Tissue Proteins/metabolism , Neurons/pathology , Simian Acquired Immunodeficiency Syndrome/metabolism , Synapses/pathology , Animals , Axons/pathology , Connexins/antagonists & inhibitors , Dendritic Spines/pathology , Gray Matter/pathology , Humans , Macaca mulatta , Nerve Tissue Proteins/antagonists & inhibitors , Virus Replication , White Matter/pathology
13.
J Neurochem ; 158(2): 429-443, 2021 07.
Article in English | MEDLINE | ID: mdl-33655498

ABSTRACT

The major barrier to eradicating Human immunodeficiency virus-1 (HIV) infection is the generation of tissue-associated quiescent long-lasting viral reservoirs refractory to therapy. Upon interruption of anti-retroviral therapy (ART), HIV replication can be reactivated. Within the brain, microglia/macrophages and a small population of astrocytes are infected with HIV. However, the role of astrocytes as a potential viral reservoir is becoming more recognized because of the improved detection and quantification of HIV viral reservoirs. In this report, we examined the infectivity of human primary astrocytes in vivo and in vitro, and their capacity to maintain HIV infection, become latently infected, be reactivated, and transfer new HIV virions into neighboring cells. Analysis of human brain tissue sections obtained from HIV-infected individuals under effective and prolonged ART indicates that a small population of astrocytes has integrated HIV-DNA. In vitro experiments using HIV-infected human primary astrocyte cultures confirmed a low percentage of astrocytes had integrated HIV-DNA, with poor to undetectable replication. Even in the absence of ART, long-term culture results in latency that could be transiently reactivated with histone deacetylase inhibitor, tumor necrosis factor-alpha (TNF-α), or methamphetamine. Reactivation resulted in poor viral production but efficient cell-to-cell viral transfer into cells that support high viral replication. Together, our data provide a new understanding of astrocytes' role as viral reservoirs within the central nervous system (CNS).


Subject(s)
Astrocytes/virology , Brain/virology , HIV Infections/pathology , HIV Infections/virology , HIV , Virus Replication/drug effects , Adult , Aged , Antiretroviral Therapy, Highly Active , Child, Preschool , DNA, Viral/genetics , Female , HIV Infections/transmission , Histone Deacetylase Inhibitors/pharmacology , Humans , Male , Methamphetamine/pharmacology , Middle Aged , Primary Cell Culture , Tumor Necrosis Factor-alpha/pharmacology
14.
J Immunol ; 205(10): 2726-2741, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33037140

ABSTRACT

HIV has become a chronic disease despite the effective use of antiretroviral therapy (ART). However, the mechanisms of tissue colonization, viral evolution, generation of viral reservoirs, and compartmentalization are still a matter of debate due to the challenges involved in examining early events of infection at the cellular and molecular level. Thus, there is still an urgent need to explore these areas to develop effective HIV cure strategies. In this study, we describe the early events of tissue colonization and compartmentalization as well as the role of tunneling nanotube-like structures during viral spread in the presence and absence of effective antiretroviral treatment. To examine these mechanisms, NOD/SCID IL-2 RG-/- humanized mice were either directly infected with HIVADA or with low numbers of HIVADA-infected leukocytes to limit tissue colonization in the presence and absence of TAK779, an effective CCR5 blocker of HIV entry. We identify that viral seeding in tissues occurs early in a tissue- and cell type-specific manner (24-72 h). Reduction in systemic HIV replication by TAK779 treatment did not affect tissue seeding or spreading, despite reduced systemic viral replication. Tissue-associated HIV-infected cells had different properties than cells in the circulation because the virus continues to spread in tissues in a tunneling nanotube-like structure-dependent manner, despite ART. Thus, understanding these mechanisms can provide new approaches to enhance the efficacy of existing ART and HIV infection cure strategies.


Subject(s)
Anti-Retroviral Agents/administration & dosage , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/virology , HIV Infections/immunology , HIV-1/pathogenicity , Amides/administration & dosage , Animals , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , HIV Infections/blood , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/immunology , HIV-1/isolation & purification , Hematopoietic Stem Cell Transplantation , Humans , Interleukin Receptor Common gamma Subunit/genetics , Mice , Mice, Knockout , Quaternary Ammonium Compounds/administration & dosage , Transplantation Chimera , Viral Load , Virus Integration/drug effects , Virus Integration/immunology , Virus Internalization/drug effects , Virus Replication/drug effects , Virus Replication/immunology
15.
iScience ; 23(9): 101450, 2020 Sep 25.
Article in English | MEDLINE | ID: mdl-32882515

ABSTRACT

Glioblastoma (GBM) is the most prevalent and aggressive tumor in the central nervous system. Surgical resection followed by concurrent radiotherapy (ionizing radiation [IR]) and temozolomide (TMZ) is the standard of care for GBM. However, a large subset of patients offer resistance or become adapted to TMZ due mainly to the DNA repair enzyme O6-methylguanine-DNA methyltransferase (MGMT). Thus, alternative mechanisms of MGMT deregulation have been proposed but are heretofore unproven. We show that heterogeneous GBM cells express tunneling nanotubes (TNTs) upon oxidative stress and TMZ/IR treatment. We identified that MGMT protein diffused from resistant to sensitive cells upon exposure to TMZ/IR, resulting in protection against cytotoxic therapy in a TNT-dependent manner. In vivo analysis of resected GBM tumors support our hypothesis that the MGMT protein, but not its mRNA, was associated with TNT biomarkers. We propose that targeting TNT formation could be an innovative strategy to overcome treatment resistance in GBM.

16.
EBioMedicine ; 51: 102503, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31806564

ABSTRACT

BACKGROUND: In developed countries, Human Immunodeficiency Virus type-1 (HIV-1) infection has become a chronic disease despite the positive effects of anti-retroviral therapies (ART), but still at least half of the HIV infected population shown signs of cognitive impairment. Therefore, biomarkers of HIV cognitive decline are urgently needed. METHODS: We analyze the opening of one of the larger channels expressed by humans, pannexin-1 (Panx-1) channels, in the uninfected and HIV infected population (n = 175). We determined channel opening and secretion of intracellular second messengers released through the channel such as PGE2 and ATP. Also, we correlated the opening of Panx-1 channels with the circulating levels of PGE2 and ATP as well as cogntive status of the individuals analyzed. FINDINGS: Here, we demonstrate that Panx-1 channels on fresh PBMCs obtained from uninfected individuals are closed and no significant amounts of PGE2 and ATP are detected in the circulation. In contrast, in all HIV-infected individuals analyzed, even the ones under effective ART, a spontaneous opening of Panx-1 channels and increased circulating levels of PGE2 and ATP were detected. Circulating levels of ATP were correlated with cognitive decline in the HIV-infected population supporting that ATP is a biomarker of cognitive disease in the HIV-infected population. INTERPRETATION: We propose that circulating levels of ATP could predict CNS compromise and lead to the breakthroughs necessary to detect and prevent brain compromise in the HIV-infected population.


Subject(s)
Adenosine Triphosphate/blood , Cognitive Dysfunction/blood , HIV Infections/blood , Adult , Aged , Biomarkers/blood , Blood-Brain Barrier/pathology , Cognitive Dysfunction/physiopathology , Connexins/blood , Dinoprostone/blood , Female , HIV Infections/physiopathology , Humans , Inflammation Mediators/blood , Ion Channel Gating , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Male , Middle Aged , Nerve Tissue Proteins/blood , Transendothelial and Transepithelial Migration
17.
J Neurochem ; 151(5): 558-569, 2019 12.
Article in English | MEDLINE | ID: mdl-31381153

ABSTRACT

Chemical and electrical synapses are the two major communication systems that permit cell-to-cell communication within the nervous system. Although most studies are focused on chemical synapses (glutamate, γ-aminobutyric acid, and other neurotransmitters), clearly both types of synapses interact and cooperate to allow the coordination of several cell functions within the nervous system. The pineal gland has limited independent axonal innervation and not every cell has access to nerve terminals. Thus, additional communication systems, such as gap junctions, have been postulated to coordinate metabolism and signaling. Using acutely isolated glands and dissociated cells, we found that gap junctions spread glycogenolytic signals from cells containing adrenoreceptors to the entire gland lacking these receptors. Our data using glycogen and lactate quantification, electrical stimulation, and high-performance liquid chromatography with electrochemical detection, demonstrate that gap junctional communication between cells of the rat pineal gland allows cell-to-cell propagation of norepinephrine-induced signal that promotes glycogenolysis throughout the entire gland. Thus, the interplay of both synapses is essential for coordinating glycogen metabolism and lactate production in the pineal gland.


Subject(s)
Cell Communication/physiology , Electrical Synapses/metabolism , Glycogenolysis/physiology , Norepinephrine/metabolism , Pineal Gland/metabolism , Animals , Female , Male , Rats , Rats, Sprague-Dawley
18.
Sci Rep ; 9(1): 3941, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30850623

ABSTRACT

Currently, a major barrier to curing HIV infection is the generation of tissue-associated, non-replicating, long-lasting viral reservoirs that are refractory to therapy and can be reactivated upon anti-retroviral therapy interruption. One of these reservoirs are latently HIV-infected macrophages. Here, we show that HIV infection of macrophages results in survival of a small population of infected cells that are metabolically altered and characterized by mitochondrial fusion, lipid accumulation, and reduced mitochondrial ATP production. No changes in glycolysis were detected. Metabolic analysis indicated an essential role of succinate and other TCA metabolites in the tricarboxylic acid (TCA) cycle in mediating lipid accumulation and oxidative phosphorylation (OXPHOS) in the mitochondria. Furthermore, we show that while uninfected and HIV infected macrophages use fatty acids and glucose as primary sources of energy, surviving HIV infected macrophages also use glutamine/glutamate as a major energy source, and blocking these new sources of energy resulted in the killing of latent HIV infected macrophages. Together, our data provide a new understanding of the formation, properties, and potential novel ways to eliminate macrophage viral reservoirs.


Subject(s)
HIV Infections/immunology , Macrophages/metabolism , Blotting, Western , Citric Acid Cycle , Energy Metabolism , Glutamic Acid/metabolism , Glutamine/metabolism , Glycolysis , HIV Infections/metabolism , Humans , Lipid Droplets/metabolism , Macrophages/virology , Mitochondria/metabolism
19.
Cancer Rep (Hoboken) ; 2(6): e1220, 2019 12.
Article in English | MEDLINE | ID: mdl-32729241

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is a highly aggressive primary brain tumor. Currently, the suggested line of action is the surgical resection followed by radiotherapy and treatment with the adjuvant temozolomide, a DNA alkylating agent. However, the ability of tumor cells to deeply infiltrate the surrounding tissue makes complete resection quite impossible, and, in consequence, the probability of tumor recurrence is high, and the prognosis is not positive. GBM is highly heterogeneous and adapts to treatment in most individuals. Nevertheless, these mechanisms of adaption are unknown. RECENT FINDINGS: In this review, we will discuss the recent discoveries in molecular and cellular heterogeneity, mechanisms of therapeutic resistance, and new technological approaches to identify new treatments for GBM. The combination of biology and computer resources allow the use of algorithms to apply artificial intelligence and machine learning approaches to identify potential therapeutic pathways and to identify new drug candidates. CONCLUSION: These new approaches will generate a better understanding of GBM pathogenesis and will result in novel treatments to reduce or block the devastating consequences of brain cancers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/therapy , Genetic Heterogeneity , Glioblastoma/therapy , Precision Medicine/methods , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain/pathology , Brain/surgery , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cancer Vaccines/administration & dosage , Chemoradiotherapy, Adjuvant/methods , Clonal Evolution , Computational Biology , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Mutational Analysis , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Drug Resistance, Neoplasm/genetics , Glioblastoma/genetics , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Machine Learning , Microtubules/drug effects , Microtubules/pathology , Models, Genetic , Mutation , Precision Medicine/trends , Prognosis , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
20.
Photochem Photobiol ; 94(6): 1092-1099, 2018 11.
Article in English | MEDLINE | ID: mdl-29964295

ABSTRACT

Naphthoxazole derivatives are small heterocyclic compounds endowed with outstanding fluorescence properties. In this work, we report a detailed study of the intense white light fluorescence observed in naphthoxazole-quinoline dyads in solvent mixtures including at least a strong hydrogen bonding solvent. The same phenomenon was also studied in inclusion complexes naphthoxazole derivatives-sulfonated-ßCD either in aqueous solution as well as in solid phase. A novel mechanism of white molecular fluorescence generation based on solvent-to-fluorophore proton transfer facilitated by ground state hydrogen bonding was characterized. The emission combines both, a blue charge transfer fluorescence emitted by the locally excited singlet state along with a red-shifted emission from a proton transfer complex.

SELECTION OF CITATIONS
SEARCH DETAIL
...