Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
2.
Cancer Res Commun ; 3(5): 917-932, 2023 05.
Article in English | MEDLINE | ID: mdl-37377887

ABSTRACT

Tumor-infiltrating lymphocytes (TIL) that can recognize and kill tumor cells have curative potential in subsets of patients treated with adoptive cell transfer (ACT). However, lack of TIL therapeutic efficacy in many patients may be due in large part to a paucity of tumor-reactive T cells in TIL and the exhausted and terminally differentiated status of those tumor-reactive T cells. We sought to reprogram exhausted TIL that possess T-cell receptors (TCR) specific for tumor antigens into induced pluripotent stem cells (iPSC) to rejuvenate them for more potent ACT. We first attempted to reprogram tumor neoantigen-specific TIL by αCD3 Ab prestimulation which resulted in failure of establishing tumor-reactive TIL-iPSCs, instead, T cell-derived iPSCs from bystander T cells were established. To selectively activate and enrich tumor-reactive T cells from the heterogenous TIL population, CD8+ PD-1+ 4-1BB+ TIL population were isolated after coculture with autologous tumor cells, followed by direct reprogramming into iPSCs. TCR sequencing analysis of the resulting iPSC clones revealed that reprogrammed TIL-iPSCs encoded TCRs that were identical to the pre-identified tumor-reactive TCRs found in minimally cultured TIL. Moreover, reprogrammed TIL-iPSCs contained rare tumor antigen-specific TCRs, which were not detectable by TCR sequencing of the starting cell population. Thus, reprogramming of PD-1+ 4-1BB+ TIL after coculture with autologous tumor cells selectively generates tumor antigen-specific TIL-iPSCs, and is a distinctive method to enrich and identify tumor antigen-specific TCRs of low frequency from TIL. Significance: Reprogramming of TIL into iPSC holds great promise for the future treatment of cancer due to their rejuvenated nature and the retention of tumor-specific TCRs. One limitation is the lack of selective and efficient methods for reprogramming tumor-specific T cells from polyclonal TIL. Here we addressed this limitation and present a method to efficiently reprogram TIL into iPSC colonies carrying diverse tumor antigen reactive TCR recombination.


Subject(s)
Induced Pluripotent Stem Cells , Neoplasms , Humans , Lymphocytes, Tumor-Infiltrating , Programmed Cell Death 1 Receptor , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Antigens, Neoplasm
3.
J Vis Exp ; (150)2019 08 09.
Article in English | MEDLINE | ID: mdl-31449236

ABSTRACT

The inheritance of pre-rearranged T cell receptors (TCRs) and their epigenetic rejuvenation make induced pluripotent stem cell (iPSC)-derived T cells a promising source for adoptive T cell therapy (ACT). However, classical in vitro methods for producing regenerated T cells from iPSC result in either innate-like or terminally differentiated T cells, which are phenotypically and functionally distinct from naïve T cells. Recently, a novel three-dimensional (3D) thymic culture system was developed to generate a homogenous subset of CD8αß+ antigen-specific T cells with a naïve T cell-like functional phenotype, including the capacity for proliferation, memory formation, and tumor suppression in vivo. This protocol avoids aberrant developmental fates, allowing for the generation of clinically relevant iPSC-derived T cells, designated as iPSC-derived thymic emigrants (iTE), while also providing a potent tool to elucidate the subsequent functions necessary for T cell maturation after thymic selection.


Subject(s)
Antigens, Neoplasm/immunology , Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/cytology , Thymus Gland/cytology , Thymus Gland/immunology , Animals , Cell Differentiation , Cell Line, Tumor , Mice , T-Lymphocytes/cytology , T-Lymphocytes/immunology
4.
J Virol ; 92(14)2018 07 15.
Article in English | MEDLINE | ID: mdl-29720520

ABSTRACT

Despite the long-standing observation that herpes simplex virus (HSV) latency-associated transcript (LAT) promoter deletion viruses show impaired recurrence phenotypes in relevant animal models, the mechanism by which these sequences exert this phenotypic effect is unknown. We constructed and evaluated four mutant HSV-2 isolates with targeted mutations in the LAT promoter and LAT-associated microRNAs (miRNAs) affecting (i) the LAT TATA box; (ii) the LAT ICP4-binding site; (iii) miRNA I (miR-I) and miR-II (miR-I/II), which both target ICP34.5; and (iv) miR-III, which targets ICP0. While the LAT TATA box mutant caused milder acute infections than wild-type (WT) virus, there was no difference in the recurrence phenotype between these viruses. LAT and miRNA expression during latency was not impaired by this mutation, suggesting that other promoter elements may be more important for latent HSV-2 LAT expression. Mutation of the LAT ICP4-binding site also did not cause an in vivo phenotypic difference between mutant and WT viruses. Acute infection and reactivation from latency of the miR-I/II mutant were similar to those of its rescuant, although the acute infection was slightly reduced in severity relative to that caused by the wild-type virus. The miR-III mutant also exhibited WT phenotypes in acute and recurrent phases of infection. While they do not rule out an effect of these elements in human latency or reactivation, these findings do not identify a specific role for LAT or LAT-associated miRNAs in the HSV-2 LAT promoter deletion phenotype in guinea pigs. Thus, other sequences in this region may play a more important role in the long-studied LAT-associated phenotype in animals.IMPORTANCE While it has been known for several decades that specific HSV-1 and HSV-2 sequences near the LAT promoter are required for efficient viral reactivation in animal models, the mechanism is still not known. We constructed four mutant viruses with the goal of identifying critical sequence elements and of specifically testing the hypothesis that microRNAs that are expressed during latency play a role. Determination that specific LAT promoter sequences and miRNA sequences do not influence viral reactivation of HSV-2 helps to narrow down the search for the mechanism by which the virus controls its latency and recurrence phenotype.


Subject(s)
Herpes Simplex/virology , Herpesvirus 2, Human/physiology , MicroRNAs/genetics , Mutation , Viral Proteins/genetics , Virus Activation/genetics , Virus Latency/genetics , Animals , Female , Gene Expression Regulation, Viral , Guinea Pigs , Herpes Simplex/genetics , Herpes Simplex/metabolism , Promoter Regions, Genetic , RNA, Viral , Vagina/virology
5.
Cell Rep ; 22(12): 3175-3190, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29562175

ABSTRACT

Induced pluripotent stem cell (iPSC)-derived T cells may provide future therapies for cancer patients, but those generated by current methods, such as the OP9/DLL1 system, have shown abnormalities that pose major barriers for clinical translation. Our data indicate that these iPSC-derived CD8 single-positive T cells are more like CD4+CD8+ double-positive T cells than mature naive T cells because they display phenotypic markers of developmental arrest and an innate-like phenotype after stimulation. We developed a 3D thymic culture system to avoid these aberrant developmental fates, generating a homogeneous subset of CD8αß+ antigen-specific T cells, designated iPSC-derived thymic emigrants (iTEs). iTEs exhibit phenotypic and functional similarities to naive T cells both in vitro and in vivo, including the capacity for expansion, memory formation, and tumor suppression. These data illustrate the limitations of current methods and provide a tool to develop the next generation of iPSC-based antigen-specific immunotherapies.


Subject(s)
Imaging, Three-Dimensional/methods , Induced Pluripotent Stem Cells/cytology , Thymus Gland/cytology , Cell Culture Techniques/methods , Cell Differentiation/physiology , Humans , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/metabolism , Thymus Gland/diagnostic imaging , Thymus Gland/immunology
6.
J Clin Invest ; 127(7): 2626-2630, 2017 Jun 30.
Article in English | MEDLINE | ID: mdl-28581445

ABSTRACT

Chronic viral infections are difficult to treat, and new approaches are needed, particularly those aimed at reducing reactivation by enhancing immune responses. Herpes simplex virus (HSV) establishes latency and reactivates frequently, and breakthrough reactivation can occur despite suppressive antiviral therapy. Virus-specific T cells are important to control HSV, and proliferation of activated T cells requires increased metabolism of glutamine. Here, we found that supplementation with oral glutamine reduced virus reactivation in latently HSV-1-infected mice and HSV-2-infected guinea pigs. Transcriptome analysis of trigeminal ganglia from latently HSV-1-infected, glutamine-treated WT mice showed upregulation of several IFN-γ-inducible genes. In contrast to WT mice, supplemental glutamine was ineffective in reducing the rate of HSV-1 reactivation in latently HSV-1-infected IFN-γ-KO mice. Mice treated with glutamine also had higher numbers of HSV-specific IFN-γ-producing CD8 T cells in latently infected ganglia. Thus, glutamine may enhance the IFN-γ-associated immune response and reduce the rate of reactivation of latent virus infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Glutamine/pharmacology , Herpes Genitalis/drug therapy , Virus Activation/drug effects , Animals , CD8-Positive T-Lymphocytes/pathology , Guinea Pigs , Herpes Genitalis/genetics , Herpes Genitalis/immunology , Herpes Genitalis/pathology , Herpesvirus 1, Human/physiology , Herpesvirus 2, Human/physiology , Interferon-gamma/genetics , Interferon-gamma/immunology , Mice , Mice, Knockout , Virus Activation/genetics , Virus Activation/immunology
7.
J Virol ; 89(9): 4837-48, 2015 May.
Article in English | MEDLINE | ID: mdl-25673716

ABSTRACT

UNLABELLED: In order to understand factors that may influence latency-associated transcription and latency-associated transcript (LAT) phenotypes, we studied the expression of the herpes simplex virus 2 (HSV-2) LAT-associated microRNAs (miRNAs). We mapped the transcription initiation sites of all three primary miRNA transcripts and identified the ICP4-binding sequences at the transcription initiation sites of both HSV-2 LAT (pri-miRNA for miR-I and miR-II, which target ICP34.5, and miR-III, which targets ICP0) and L/ST (a pri-miRNA for miR-I and miR-II) but not at that of the primary miR-H6 (for which the target is unknown). We confirmed activity of the putative HSV-2 L/ST promoter and found that ICP4 trans-activates the L/ST promoter when the ICP4-binding site at its transcription initiation site is mutated, suggesting that ICP4 may play a dual role in regulating transcription of L/ST and, consequently, of miR-I and miR-II. LAT exon 1 (containing LAT enhancer sequences), together with the LAT promoter region, comprises a bidirectional promoter required for the expression of both LAT-encoded miRNAs and miR-H6 in latently infected mouse ganglia. The ability of ICP4 to suppress ICP34.5-targeting miRNAs and to activate lytic viral genes suggests that ICP4 could play a key role in the switch between latency and reactivation. IMPORTANCE: The HSV-2 LAT and viral miRNAs expressed in the LAT region are the most abundant viral transcripts during HSV latency. The balance between the expression of LAT and LAT-associated miRNAs and the expression of lytic viral transcripts from the opposite strand appears to influence whether individual HSV-infected neurons will be latently or productively infected. The outcome of neuronal infection may thus depend on regulation of gene expression of the corresponding primary miRNAs. In the present study, we characterize promoter sequences responsible for miRNA expression, including identification of the primary miRNA 5' ends and evaluation of ICP4 response. These findings provide further insight into the virus' strategy to tightly control expression of lytic cycle genes (especially the neurovirulence factor, ICP34.5) and suggest a mechanism (via ICP4) for the transition from latency to reactivated productive infection.


Subject(s)
Gene Expression Regulation, Viral , Herpesvirus 2, Human/genetics , MicroRNAs/biosynthesis , Animals , Binding Sites , Female , Gene Expression Profiling , Mice , Promoter Regions, Genetic , Protein Binding , Transcription Factors/metabolism , Transcription Initiation Site
8.
Sci Transl Med ; 6(265): 265ra169, 2014 Dec 03.
Article in English | MEDLINE | ID: mdl-25473037

ABSTRACT

Herpesviruses are highly prevalent and maintain lifelong latent reservoirs, thus posing challenges to the control of herpetic disease despite the availability of antiviral pharmaceuticals that target viral DNA replication. The initiation of herpes simplex virus infection and reactivation from latency is dependent on a transcriptional coactivator complex that contains two required histone demethylases, LSD1 (lysine-specific demethylase 1) and a member of the JMJD2 family (Jumonji C domain-containing protein 2). Inhibition of either of these enzymes results in heterochromatic suppression of the viral genome and blocks infection and reactivation in vitro. We demonstrate that viral infection can be epigenetically suppressed in three animal models of herpes simplex virus infection and disease. Treating animals with the monoamine oxidase inhibitor tranylcypromine to inhibit LSD1 suppressed viral lytic infection, subclinical shedding, and reactivation from latency in vivo. This phenotypic suppression was correlated with enhanced epigenetic suppression of the viral genome and suggests that, even during latency, the chromatin state of the virus is dynamic. Therefore, epi-pharmaceuticals may represent a promising approach to treat herpetic diseases.


Subject(s)
Epigenesis, Genetic , Herpesviridae Infections/metabolism , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Oxidoreductases, N-Demethylating/physiology , Animals , Disease Models, Animal , Female , Genome, Viral , Guinea Pigs , Histone Demethylases , Mice , Mice, Inbred BALB C , Monoamine Oxidase Inhibitors/chemistry , Phenotype , Protein Structure, Tertiary , Rabbits , Recurrence , Tranylcypromine/chemistry , Vagina/virology , Virus Activation , Virus Latency , Virus Replication/drug effects , Virus Shedding
9.
Transfusion ; 54(6): 1604-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24410697

ABSTRACT

BACKGROUND: Bacterial sepsis is still a complication in patients transfused with stored platelets (PLTs). We have recently demonstrated that selected antimicrobial peptides (AMPs) have bactericidal activity in bacteria-spiked PLTs. In a subsequent preclinical study, we have also shown that these AMPs do not elicit antibody response in rabbits and treatment of PLTs before transfusion does not affect their in vivo recovery and survival in severe combined immunodeficient mice. Here we have selected two such AMPs, Arg-Trp (RW) repeats of tri- and tetra-peptides (RW3 and RW4) in combination (i.e., cocktail), and evaluated their effect on the in vitro properties of PLTs. STUDY DESIGN AND METHODS: Leukoreduced ABO- and D-identical whole blood-derived PLT concentrates were pooled and divided into two 60-mL aliquots in CLX storage bags. On Day 0, one bag received a peptide cocktail of RW3 plus RW4 at 0.01 mmol/L final concentration (test) and the other bag received only phosphate-buffered saline (PBS), the AMP solvent (control). The treated PLTs were stored for 7 days at 20 to 24°C. Samples were collected on Days 1, 5, and 7 to evaluate the in vitro properties of PLTs with standard assays. RESULTS: In vitro properties of the RW3 plus RW4 cocktail-treated PLTs were similar to those incubated with PBS only. There were no significant differences between the control and test PLTs during the 7-day storage. CONCLUSION: Leukoreduced whole blood-derived PLTs treated with a mixture of RW3 and RW4 peptides maintain their in vitro properties during 7 days of storage.


Subject(s)
Anti-Infective Agents/pharmacology , Blood Platelets/drug effects , Leukocyte Reduction Procedures , Blood Preservation/methods , Humans , Platelet Transfusion
10.
Transfusion ; 54(3): 569-76, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23808572

ABSTRACT

BACKGROUND: Bacterial sepsis is a complication attributed to room temperature (RT)-stored platelets (PLTs) in transfusion medicine. Antimicrobial peptides (AMPs) are emerging as new therapeutic agents against microbes. We had previously demonstrated bactericidal activity of select synthetic AMPs against six types of bacteria in stored PLTs. In this report, we tested these AMPs for their potential antibody response and interference with the recovery and survival of human PLTs in an animal model. STUDY DESIGN AND METHODS: Two separate studies were conducted to evaluate the safety of the synthetic AMPs. 1) Two AMPs (PD3 and PD4), derived from thrombin-induced human PLT microbicidal protein, and four repeats of arginine-tryptophan (RW), containing two to five repeats (RW2-RW5), were tested in rabbits for potential antibody response. 2) RT-stored human PLTs treated for 2 hours with each of the six AMPs individually or with phosphate-buffered saline (PBS) alone were infused into severe combined immunodeficient (SCID) mice to evaluate their in vivo recovery and survival by flow cytometry. RESULTS: Except for PD3, which showed a weak immune response, all other peptides did not induce any detectable antibodies in rabbits. Furthermore, all six AMPs tested did not significantly affect the in vivo recovery and survival of human PLTs in SCID mice compared to PBS alone-treated PLTs. CONCLUSION: Preclinical evaluation studies reported here demonstrate that the selected AMPs used in the study did not adversely affect the human PLT recovery and survival in the SCID mouse model, suggesting further study of AMPs toward addressing the bacterial contamination of PLTs.


Subject(s)
Anti-Infective Agents/pharmacology , Blood Platelets/drug effects , Blood Preservation/methods , Animals , Antimicrobial Cationic Peptides/pharmacology , Flow Cytometry , Humans , Mice , Mice, SCID , Rabbits
11.
Proc Natl Acad Sci U S A ; 108(12): 4986-90, 2011 Mar 22.
Article in English | MEDLINE | ID: mdl-21383158

ABSTRACT

Nitric oxide (NO) is a signaling molecule that can trigger adaptive (physiological) or maladaptive (pathological) responses to stress stimuli in a context-dependent manner. We have previously reported that NO may signal axonal injury to neighboring glial cells. In this study, we show that mice deficient in neuronal nitric oxide synthase (nNOS-/-) are more vulnerable than WT mice to toxin-induced peripheral neuropathy. The administration of NO donors to primary dorsal root ganglion cultures prevents axonal degeneration induced by acrylamide in a dose-dependent manner. We demonstrate that NO-induced axonal protection is dependent on hypoxia-inducible factor (HIF)-1-mediated transcription of erythropoietin (EPO) within glial (Schwann) cells present in the cultures. Transduction of Schwann cells with adenovirus AdCA5 encoding a constitutively active form of HIF-1α results in amelioration of acrylamide-induced axonal degeneration in an EPO-dependent manner. Mice that are partially deficient in HIF-1α (HIF-1α+/-) are also more susceptible than WT littermates to toxic neuropathy. Our results indicate that NO→HIF-1→EPO signaling represents an adaptive mechanism that protects against axonal degeneration.


Subject(s)
Axons/metabolism , Erythropoietin/biosynthesis , Hypoxia-Inducible Factor 1/metabolism , Neurodegenerative Diseases/metabolism , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide/metabolism , Adenoviridae , Animals , Axons/pathology , Dose-Response Relationship, Drug , Erythropoietin/genetics , Ganglia, Spinal/metabolism , Hypoxia-Inducible Factor 1/genetics , Mice , Mice, Knockout , Neurodegenerative Diseases/genetics , Nitric Oxide/genetics , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type I/genetics , Rats , Rats, Sprague-Dawley , Schwann Cells/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Transduction, Genetic
12.
Hum Mol Genet ; 20(9): 1844-53, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21325354

ABSTRACT

Spinal muscular atrophy (SMA) is an inherited motor neuron disease caused by the mutation of the survival motor neuron 1 (SMN1) gene and deficiency of the SMN protein. Severe SMA mice have abnormal motor function and small, immature myofibers early in development suggesting that SMN protein deficiency results in retarded muscle growth. Insulin-like growth factor 1 (IGF-1) stimulates myoblast proliferation, induces myogenic differentiation and generates myocyte hypertrophy in vitro and in vivo. We hypothesized that increased expression of IGF-1 specifically in skeletal muscle would attenuate disease features of SMAΔ7 mice. SMAΔ7 mice overexpressing a local isoform of IGF-1 (mIGF-1) in muscle showed enlarged myofibers and a 40% increase in median survival compared with mIGF-1-negative SMA littermates (median survival = 14 versus 10 days, respectively, log-rank P = 0.025). Surprisingly, this was not associated with a significant improvement in motor behavior. Treatment of both mIGF-1(NEG) and mIGF-1(POS) SMA mice with the histone deacetylase inhibitor, trichostatin A (TSA), resulted in a further extension of survival and improved motor behavior, but the combination of mIGF-1 and TSA treatment was not synergistic. These results show that increased mIGF-1 expression restricted to muscle can modulate the phenotype of SMA mice indicating that therapeutics targeted to muscle alone should not be discounted as potential disease-modifying therapies in SMA. IGF-1 may warrant further investigation in mild SMA animal models and perhaps SMA patients.


Subject(s)
Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/metabolism , Up-Regulation , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Knockout , Motor Activity , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/physiopathology , SMN Complex Proteins/genetics , SMN Complex Proteins/metabolism
13.
Am J Physiol Gastrointest Liver Physiol ; 299(4): G833-43, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20689059

ABSTRACT

Acute lung injury (ALI) and the development of the multiple organ dysfunction syndrome (MODS) are major causes of death in trauma patients. Gut inflammation and loss of gut barrier function as a consequence of splanchnic ischemia-reperfusion (I/R) have been implicated as the initial triggering events that contribute to the development of the systemic inflammatory response, ALI, and MODS. Since hypoxia-inducible factor (HIF-1) is a key regulator of the physiological and pathophysiological response to hypoxia, we asked whether HIF-1 plays a proximal role in the induction of gut injury and subsequent lung injury. Utilizing partially HIF-1α-deficient mice in a global trauma hemorrhagic shock (T/HS) model, we found that HIF-1 activation was necessary for the development of gut injury and that the prevention of gut injury was associated with an abrogation of lung injury. Specifically, in vivo studies demonstrated that partial HIF-1α deficiency ameliorated T/HS-induced increases in intestinal permeability, bacterial translocation, and caspase-3 activation. Lastly, partial HIF-1α deficiency reduced TNF-α, IL-1ß, cyclooxygenase-2, and inducible nitric oxide synthase levels in the ileal mucosa after T/HS whereas IL-1ß mRNA levels were reduced in the lung after T/HS. This study indicates that prolonged intestinal HIF-1 activation is a proximal regulator of I/R-induced gut mucosal injury and gut-induced lung injury. Consequently, these results provide unique information on the initiating events in trauma-hemorrhagic shock-induced ALI and MODS as well as potential therapeutic insights.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Inflammation/metabolism , Intestinal Diseases/metabolism , Intestines/injuries , Reperfusion Injury/metabolism , Animals , Apoptosis , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation/physiology , Genotype , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intestinal Diseases/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestines/pathology , Lung/metabolism , Lung/pathology , Lung Injury/metabolism , Lung Injury/pathology , Mice , Permeability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reperfusion Injury/pathology , Shock, Hemorrhagic/metabolism , Shock, Hemorrhagic/pathology
14.
Proc Natl Acad Sci U S A ; 106(44): 18769-74, 2009 Nov 03.
Article in English | MEDLINE | ID: mdl-19841279

ABSTRACT

Diabetes is a major risk factor for ischemic disease. Treatment options for diabetic patients with peripheral arterial disease when revascularization is not possible are limited, resulting in a high incidence of limb amputation. We evaluated the therapeutic potential of AdCA5, an adenovirus encoding a constitutively active form of HIF-1alpha, in a diabetic model of critical limb ischemia. Diabetic db/db and nondiabetic db/+ mice were subjected to unilateral femoral artery ligation. Limb perfusion, tissue viability, and motor function were more severely impaired in db/db mice. Intramuscular injection of AdCA5 into the ischemic limb of db/db mice increased the recovery of limb perfusion and function, reduced tissue necrosis, rescued the diabetes-associated impairment of circulating angiogenic cells, enhanced endothelial nitric oxide synthase activation, and increased vessel density and luminal area in the ischemic limb.


Subject(s)
Adenoviridae/genetics , Diabetes Mellitus, Experimental/complications , Extremities/blood supply , Genetic Therapy , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/therapeutic use , Ischemia/therapy , Animals , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Extremities/pathology , Femoral Artery/surgery , Humans , Immunohistochemistry , Ischemia/complications , Ischemia/enzymology , Ischemia/pathology , Ligation , Mice , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Neovascularization, Physiologic , Nitric Oxide Synthase Type III/metabolism , Signal Transduction
15.
J Neurosci ; 29(3): 842-51, 2009 Jan 21.
Article in English | MEDLINE | ID: mdl-19158308

ABSTRACT

The motor neuron disease spinal muscular atrophy (SMA) causes profound muscle weakness that most often leads to early death. At autopsy, SMA is characterized by loss of motor neurons and muscle atrophy, but the initial cellular events that precipitate motor unit dysfunction and loss remain poorly characterized. Here, we examined the function and corresponding structure of neuromuscular junction (NMJ) synapses in a mouse model of severe SMA (hSMN2/delta7SMN/mSmn-/-). Surprisingly, most SMA NMJs remained innervated even late in the disease course; however they showed abnormal synaptic transmission. There was a two-fold reduction in the amplitudes of the evoked endplate currents (EPCs), but normal spontaneous miniature EPC (MEPC) amplitudes. These features in combination indicate reduced quantal content. SMA NMJs also demonstrated increased facilitation suggesting a reduced probability of vesicle release. By electron microscopy, we found a decreased density of synaptic vesicles that is likely to contribute to the reduced release probability. In addition to presynaptic defects, there were postsynaptic abnormalities. EPC and MEPC decay time constants were prolonged because of a slowed switch from the fetal acetylcholine receptor (AChR) gamma-subunit to the adult epsilon-subunit. There was also reduced size of AChR clusters and small myofibers, which expressed an immature pattern of myosin heavy chains. Together these results indicate that impaired synaptic vesicle release at NMJs in severe SMA is likely to contribute to failed postnatal maturation of motor units and muscle weakness.


Subject(s)
Muscular Atrophy, Spinal/pathology , Neuromuscular Junction/immunology , Neuromuscular Junction/physiopathology , Synaptic Vesicles/metabolism , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Autonomic Denervation/methods , Disease Models, Animal , Electric Stimulation , Mice , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Miniature Postsynaptic Potentials/physiology , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/genetics , Neuromuscular Junction/ultrastructure , Receptors, Cholinergic/metabolism , Survival of Motor Neuron 1 Protein/genetics , Synaptic Vesicles/ultrastructure
16.
Ann Neurol ; 64(4): 465-70, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18661558

ABSTRACT

Early treatment with the histone deacetylase inhibitor, trichostatin A, plus nutritional support extended median survival of spinal muscular atrophy mice by 170%. Treated mice continued to gain weight, maintained stable motor function, and retained intact neuromuscular junctions long after trichostatin A was discontinued. In many cases, ultimate decline of mice appeared to result from vascular necrosis, raising the possibility that vascular dysfunction is part of the clinical spectrum of severe spinal muscular atrophy. Early spinal muscular atrophy disease detection and treatment initiation combined with aggressive ancillary care may be integral to the optimization of histone deacetylase inhibitor treatment in human patients.


Subject(s)
Enzyme Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Muscular Atrophy, Spinal/therapy , Nutritional Support/methods , Age Factors , Animals , Animals, Newborn , Body Weight/drug effects , Body Weight/physiology , Disease Models, Animal , Disease Progression , Mice , Mice, Transgenic , Motor Activity/drug effects , Necrosis , Survival Analysis , Survival of Motor Neuron 1 Protein/genetics
17.
J Biol Chem ; 283(16): 10892-903, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18281291

ABSTRACT

Autophagy is a process by which cytoplasmic organelles can be catabolized either to remove defective structures or as a means of providing macromolecules for energy generation under conditions of nutrient starvation. In this study we demonstrate that mitochondrial autophagy is induced by hypoxia, that this process requires the hypoxia-dependent factor-1-dependent expression of BNIP3 and the constitutive expression of Beclin-1 and Atg5, and that in cells subjected to prolonged hypoxia, mitochondrial autophagy is an adaptive metabolic response which is necessary to prevent increased levels of reactive oxygen species and cell death.


Subject(s)
Autophagy , Hypoxia-Inducible Factor 1/metabolism , Hypoxia , Mitochondria/metabolism , Animals , Apoptosis Regulatory Proteins , Autophagy-Related Protein 5 , Beclin-1 , Cell Death , Cytoplasm/metabolism , Membrane Proteins/biosynthesis , Mice , Mice, Knockout , Microtubule-Associated Proteins/biosynthesis , Mitochondrial Proteins/biosynthesis , Models, Biological , Molecular Conformation , Proteins/metabolism , Reactive Oxygen Species
18.
Cardiovasc Res ; 77(3): 463-70, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18006459

ABSTRACT

AIMS: We investigated whether hypoxia-inducible factor 1 alpha (HIF-1 alpha) plays a role in the acute phase of ischaemic preconditioning (IPC). METHODS AND RESULTS: Hearts from wild-type (WT) mice and mice heterozygous for a null allele at the locus encoding HIF-1 alpha (HET) were subjected to IPC (10-min ischaemia/5 min reperfusion, or two cycles of 5 min ischaemia/5 min reperfusion), followed by 30 min ischaemia and reperfusion. Left ventricular-developed pressure, heart rate, and coronary flow rate were measured continuously. Apoptosis and infarct size were assessed by TUNEL assay, cleaved caspase 3 immunohistochemistry, and triphenyltetrazolium chloride staining. Production of reactive oxygen species (ROS) in isolated cardiac mitochondria was measured by a chemiluminescence assay. The phosphatase and tensin homologue (PTEN) and AKT (protein kinase B) were analysed by immunoblot assay. IPC improved functional recovery and limited infarct size and apoptosis after prolonged ischaemia-reperfusion in WT hearts, but not in HET hearts. Mitochondrial ROS production, PTEN oxidation, and AKT phosphorylation were impaired in HET hearts. WT and HET hearts were protected by adenosine, which acts via an ROS-independent mechanism. CONCLUSION: HIF-1 alpha is required for IPC-induced mitochondrial ROS production and myocardial protection against ischaemia-reperfusion injury.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Ischemic Preconditioning, Myocardial , Myocardial Infarction/prevention & control , Adenosine/pharmacology , Animals , Apoptosis , Male , Mice , Myocardial Reperfusion Injury/prevention & control , PTEN Phosphohydrolase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism
19.
Circ Res ; 101(12): 1310-8, 2007 Dec 07.
Article in English | MEDLINE | ID: mdl-17932327

ABSTRACT

Ischemia is a stimulus for production of angiogenic cytokines that activate local vascular cells and mobilize angiogenic cells to the circulation. These responses are impaired in elderly patients with peripheral arterial disease. Hypoxia-inducible factor (HIF)-1 mediates adaptive responses to ischemia, including production of angiogenic cytokines. In this study, we demonstrate that aging and HIF-1 loss-of-function impair the expression of multiple angiogenic cytokines, mobilization of angiogenic cells, maintenance of tissue viability, and recovery of limb perfusion following femoral artery ligation. We show that HIF-1 directly activates transcription of the gene encoding stem cell factor and that mice lacking the cognate receptor C-KIT have impaired recovery from ischemia. Administration of AdCA5, an adenovirus encoding a constitutively active form of HIF-1alpha, improved the recovery of perfusion in older mice to levels similar to those in young mice. Injection of AdCA5 into nonischemic limb was sufficient to increase the number of circulating angiogenic cells. These results indicate that HIF-1 activity is necessary and sufficient for the mobilization of angiogenic cells and that HIF-1alpha gene therapy can counteract the pathological effects of aging in a mouse model of limb ischemia.


Subject(s)
Aging/metabolism , Cell Movement/physiology , Hypoxia-Inducible Factor 1/metabolism , Ischemia/genetics , Ischemia/therapy , Lower Extremity/blood supply , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/therapy , Aging/genetics , Aging/pathology , Animals , Cell Movement/genetics , Cells, Cultured , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/therapeutic use , Ischemia/metabolism , Ischemia/pathology , Lower Extremity/physiology , Male , Mice , Mice, Knockout , Mice, Transgenic , Neovascularization, Pathologic/metabolism , Reperfusion/methods
20.
J Bone Miner Res ; 22(3): 366-74, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17181398

ABSTRACT

UNLABELLED: HIF-1alpha activates genes under hypoxia and was hypothesized to regulate bone regeneration. Surprisingly, HIF-1alpha+/- fracture calluses are larger, stronger, and stiffer than HIF-1alpha+/+ calluses because of decreased apoptosis. These data identify apoptosis inhibition as a means to enhance bone regeneration. INTRODUCTION: Bone regeneration subsequent to fracture involves the synergistic activation of multiple signaling pathways. Localized hypoxia after fracture activates hypoxia-inducible factor 1alpha (HIF-1alpha), leading to increased expression of HIF-1 target genes. We therefore hypothesized that HIF-1alpha is a key regulator of bone regeneration. MATERIALS AND METHODS: Fixed femoral fractures were generated in mice with partial HIF-1alpha deficiency (HIF-1alpha+/-) and wildtype littermates (HIF-1alpha+/+). Fracture calluses and intact contralateral femurs from postfracture days (PFDs) 21 and 28 (N=5-10) were subjected to microCT evaluation and four-point bending to assess morphometric and mechanical properties. Molecular analyses were carried out on PFD 7, 10, and 14 samples (N=3) to determine differential gene expression at both mRNA and protein levels. Finally, TUNEL staining was performed on PFD 14 samples (N=2) to elucidate differential apoptosis. RESULTS: Surprisingly, fracture calluses from HIF-1alpha+/- mice exhibited greater mineralization and were larger, stronger, and stiffer. Microarray analyses focused on hypoxia-induced genes revealed differential expression (between genotypes) of several genes associated with the apoptotic pathway. Real-time PCR confirmed these results, showing higher expression of proapoptotic protein phosphatase 2a (PP2A) and lower expression of anti-apoptotic B-cell leukemia/lymphoma 2 (BCL2) in HIF-1alpha+/+ calluses. Subsequent TUNEL staining showed that HIF-1alpha+/+ calluses contained larger numbers of TUNEL+ chondrocytes and osteoblasts than HIF-1alpha+/- calluses. CONCLUSIONS: We conclude that partial HIF-1alpha deficiency results in decreased chondrocytic and osteoblastic apoptosis, thereby allowing the development of larger, stiffer calluses and enhancing bone regeneration. Furthermore, apoptosis inhibition may be a promising target for developing new treatments to accelerate bone regeneration.


Subject(s)
Apoptosis , Bone Regeneration , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Osteoblasts/metabolism , Animals , Animals, Genetically Modified , Apoptosis/genetics , Bone Regeneration/genetics , Bony Callus/metabolism , Chondrocytes , Female , Femoral Fractures/genetics , Femoral Fractures/metabolism , Gene Expression Profiling , Gene Expression Regulation/genetics , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Oligonucleotide Array Sequence Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...