Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
2.
J Biol Chem ; 300(2): 105633, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38199567

ABSTRACT

Cytochrome P450 3A4 and 2D6 (EC 1.14.13.97 and 1.14.14.1; CYP3A4 and 2D6) are heme-containing enzymes that catalyze the oxidation of a wide number of xenobiotic and drug substrates and thus broadly impact human biology and pharmacologic therapies. Although their activities are directly proportional to their heme contents, little is known about the cellular heme delivery and insertion processes that enable their maturation to functional form. We investigated the potential involvement of GAPDH and chaperone Hsp90, based on our previous studies linking these proteins to intracellular heme allocation. We studied heme delivery and insertion into CYP3A4 and 2D6 after they were transiently expressed in HEK293T and GlyA CHO cells or when naturally expressed in HEPG2 cells in response to rifampicin, and also investigated their associations with GAPDH and Hsp90 in cells. The results indicate that GAPDH and its heme binding function is involved in delivery of mitochondria-generated heme to apo-CYP3A4 and 2D6, and that cell chaperone Hsp90 is additionally involved in driving their heme insertions. Uncovering how cells allocate heme to CYP3A4 and 2D6 provides new insight on their maturation processes and how this may help to regulate their functions in health and disease.


Subject(s)
Cytochrome P-450 CYP3A , Heme , Animals , Cricetinae , Humans , Cricetulus , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , HEK293 Cells , Heme/metabolism , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Oxidation-Reduction
3.
FASEB J ; 38(6): e23572, 2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38512139

ABSTRACT

Asthma is characterized by airway remodeling and hyperreactivity. Our earlier studies determined that the nitric oxide (NO)-soluble guanylyl cyclase (sGC)-cGMP pathway plays a significant role in human lung bronchodilation. However, this bronchodilation is dysfunctional in asthma due to high NO levels, which cause sGC to become heme-free and desensitized to its natural activator, NO. In order to determine how asthma impacts the various lung segments/lobes, we mapped the inflammatory regions of lungs to determine whether such regions coincided with molecular signatures of sGC dysfunction. We demonstrate using murine models of asthma (OVA and CFA/HDM) that the inflamed segments of these murine lungs can be tracked by upregulated expression of HO1 and these regions in turn overlap with regions of heme-free sGC as evidenced by a decreased sGC-α1ß1 heterodimer and an increased response to heme-independent sGC activator, BAY 60-2770, relative to naïve uninflamed regions. We also find that NO generated from iNOS upregulation in the inflamed segments has a higher impact on developing heme-free sGC as increasing iNOS activity correlates linearly with elevated heme-independent sGC activation. This excess NO works by affecting the epithelial lung hemoglobin (Hb) to become heme-free in asthma, thereby causing the Hb to lose its NO scavenging function and exposing the underlying smooth muscle sGC to excess NO, which in turn becomes heme-free. Recognition of these specific lung segments enhances our understanding of the inflamed lungs in asthma with the ultimate aim to evaluate potential therapies and suggest that regional and not global inflammation impacts lung function in asthma.


Subject(s)
Asthma , Heme Oxygenase-1 , Heme , Animals , Humans , Mice , Allergens , Heme Oxygenase-1/metabolism , Inflammation , Nitric Oxide , Soluble Guanylyl Cyclase
4.
Bioessays ; 45(8): e2300055, 2023 08.
Article in English | MEDLINE | ID: mdl-37276366

ABSTRACT

A natural heme deficiency that exists in cells outside of the circulation broadly compromises the heme contents and functions of heme proteins in cells and tissues. Recently, we found that the signaling molecule, nitric oxide (NO), can trigger or repress the deployment of intracellular heme in a concentration-dependent hormetic manner. This uncovers a new role for NO and sets the stage for it to shape numerous biological processes by controlling heme deployment and consequent heme protein functions in biology.


Subject(s)
Hemeproteins , Nitric Oxide , Nitric Oxide/metabolism , Heme/metabolism , Signal Transduction , Biology
5.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35046034

ABSTRACT

Nitric oxide (NO) signaling in biology relies on its activating cyclic guanosine monophosphate (cGMP) production by the NO receptor soluble guanylyl cyclase (sGC). sGC must obtain heme and form a heterodimer to become functional, but paradoxically often exists as an immature heme-free form in cells and tissues. Based on our previous finding that NO can drive sGC maturation, we investigated its basis by utilizing a fluorescent sGC construct whose heme level can be monitored in living cells. We found that NO generated at physiologic levels quickly triggered cells to mobilize heme to immature sGC. This occurred when NO was generated within cells or by neighboring cells, began within seconds of NO exposure, and led cells to construct sGC heterodimers and thus increase their active sGC level by several-fold. The NO-triggered heme deployment involved cellular glyceraldehyde-3-phosphate dehydrogenase (GAPDH)-heme complexes and required the chaperone hsp90, and the newly formed sGC heterodimers remained functional long after NO generation had ceased. We conclude that NO at physiologic levels triggers assembly of its own receptor by causing a rapid deployment of cellular heme. Redirecting cellular heme in response to NO is a way for cells and tissues to modulate their cGMP signaling and to more generally tune their hemeprotein activities wherever NO biosynthesis takes place.


Subject(s)
Heme/metabolism , Nitric Oxide/metabolism , Receptors, Cell Surface/metabolism , Animals , Calcium , Cyclic GMP , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Guanylate Cyclase/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Mice , Models, Biological , Protein Binding , Protein Multimerization , Protein Transport , Signal Transduction , Soluble Guanylyl Cyclase/metabolism
6.
J Biol Chem ; 299(6): 104753, 2023 06.
Article in English | MEDLINE | ID: mdl-37116709

ABSTRACT

Indoleamine-2, 3-dioxygenase (IDO1) and Tryptophan-2, 3-dioxygenase (TDO) catalyze the conversion of L-tryptophan to N-formyl-kynurenine and thus play primary roles in metabolism, inflammation, and tumor immune surveillance. Because their activities depend on their heme contents, which vary in biological settings and go up or down in a dynamic manner, we studied how their heme levels may be impacted by nitric oxide (NO) in mammalian cells. We utilized cells expressing TDO or IDO1 either naturally or via transfection and determined their activities, heme contents, and expression levels as a function of NO exposure. We found NO has a bimodal effect: a narrow range of low NO exposure promoted cells to allocate heme into the heme-free TDO and IDO1 populations and consequently boosted their heme contents and activities 4- to 6-fold, while beyond this range the NO exposure transitioned to have a negative impact on their heme contents and activities. NO did not alter dioxygenase protein expression levels, and its bimodal impact was observed when NO was released by a chemical donor or was generated naturally by immune-stimulated macrophage cells. NO-driven heme allocations to IDO1 and TDO required participation of a GAPDH-heme complex and for IDO1 required chaperone Hsp90 activity. Thus, cells can up- or downregulate their IDO1 and TDO activities through a bimodal control of heme allocation by NO. This mechanism has important biomedical implications and helps explain why the IDO1 and TDO activities in animals go up and down in response to immune stimulation.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase , Nitric Oxide , Tryptophan Oxygenase , Animals , Heme/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mammals/metabolism , Tryptophan/metabolism , Tryptophan Oxygenase/chemistry , Tryptophan Oxygenase/metabolism
7.
Mol Pharmacol ; 103(5): 286-296, 2023 05.
Article in English | MEDLINE | ID: mdl-36868790

ABSTRACT

Nitric oxide (NO)-unresponsive forms of soluble guanylyl cyclase (sGC) exist naturally and in disease can disable NO-sGC-cGMP signaling. Agonists like BAY58-2667 (BAY58) target these sGC forms, but their mechanisms of action in living cells are unclear. We studied rat lung fibroblast-6 cells and human airway smooth muscle cells that naturally express sGC and HEK293 cells that we transfected to express sGC and variants. Cells were cultured to build up different forms of sGC, and we used fluorescence and FRET-based measures to monitor BAY58-driven cGMP production and any protein partner exchange or heme loss events that may occur for each sGC species. We found that: (i) BAY58 activated cGMP production by the apo-sGCß-Hsp90 species after a 5-8 minute delay that was associated with apo-sGCß exchanging its Hsp90 partner with an sGCα subunit. (ii) In cells containing an artificially constructed heme-free sGC heterodimer, BAY58 initiated an immediate and three times faster cGMP production. However, this behavior was not observed in cells expressing native sGC under any condition. (iii) BAY58 activated cGMP production by ferric heme sGC only after a 30-minute delay, coincident with it initiating a delayed, slow ferric heme loss from sGCß We conclude that the kinetics favor BAY58 activation of the apo-sGCß-Hsp90 species over the ferric heme sGC species in living cells. The protein partner exchange events driven by BAY58 account for the initial delay in cGMP production and also limit the speed of subsequent cGMP production in the cells. Our findings clarify how agonists like BAY58 may activate sGC in health and disease. SIGNIFICANCE STATEMENT: A class of agonists can activate cyclic guanosine monophosphate (cGMP) synthesis by forms of soluble guanylyl cyclase (sGC) that do not respond to NO and accumulate in disease, but the mechanisms of action are unclear. This study clarifies what forms of sGC exist in living cells, which of these can be activated by the agonists, and the mechanisms and kinetics by which each form is activated. This information may help to hasten deployment of these agonists for pharmaceutical intervention and clinical therapy.


Subject(s)
Heme , Signal Transduction , Rats , Animals , Humans , Soluble Guanylyl Cyclase/metabolism , Heme/metabolism , HEK293 Cells , Nitric Oxide/metabolism , Heat-Shock Proteins/metabolism , Guanylate Cyclase/metabolism , Cyclic GMP/metabolism
8.
FASEB J ; 36(2): e22099, 2022 02.
Article in English | MEDLINE | ID: mdl-34972240

ABSTRACT

GAPDH, a heme chaperone, has been previously implicated in the incorporation of heme into iNOS and soluble guanylyl cyclase (sGC). Since sGC is critical for myoglobin (Mb) heme-maturation, we investigated the role of GAPDH in the maturation of this globin, as well as hemoglobins α, ß, and γ. Utilizing cell culture systems, we found that overexpression of wild-type GAPDH increased, whereas GAPDH mutants H53A and K227A decreased, the heme content of Mb and Hbα and Hbß. Overexpression of wild-type GAPDH fully recovered the heme-maturation inhibition observed with the GAPDH mutants. Partial rescue was observed by overexpression of sGCß1 but not by overexpression of a sGCΔß1 deletion mutant, which is unable to bind the sGCα1 subunit required to form the active sGCα1ß1 complex. Wild type and mutant GAPDH was found to be associated in a complex with each of the globins and Hsp90. GAPDH at endogenous levels was found to be associated with Mb in differentiating C2C12 myoblasts, and with Hbγ or Hbα in differentiating HiDEP-1 erythroid progenitor cells. Knockdown of GAPDH in C2C12 cells suppressed Mb heme-maturation. GAPDH knockdown in K562 erythroleukemia cells suppressed Hbα and Hbγ heme-maturation as well as Hb dimerization. Globin heme incorporation was not only dependent on elevated sGCα1ß1 heterodimer formation, but also influenced by iron provision and magnitude of expression of GAPDH, d-aminolevulinic acid, and FLVCR1b. Together, our data support an important role for GAPDH in the maturation of myoglobin and γ, ß, and α hemoglobins.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Heme/metabolism , Hemoglobins/metabolism , Molecular Chaperones/metabolism , Myoglobin/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , HEK293 Cells , Heme/genetics , Hemoglobins/genetics , Humans , K562 Cells , Molecular Chaperones/genetics , Mutation, Missense , Myoglobin/genetics , Sarcoglycans/genetics , Sarcoglycans/metabolism
9.
Proc Natl Acad Sci U S A ; 117(17): 9497-9507, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32300005

ABSTRACT

Nitric oxide (NO) produced by endothelial nitric oxide synthase (eNOS) is a critical mediator of vascular function. eNOS is tightly regulated at various levels, including transcription, co- and posttranslational modifications, and by various protein-protein interactions. Using stable isotope labeling with amino acids in cell culture (SILAC) and mass spectrometry (MS), we identified several eNOS interactors, including the protein plasminogen activator inhibitor-1 (PAI-1). In cultured human umbilical vein endothelial cells (HUVECs), PAI-1 and eNOS colocalize and proximity ligation assays demonstrate a protein-protein interaction between PAI-1 and eNOS. Knockdown of PAI-1 or eNOS eliminates the proximity ligation assay (PLA) signal in endothelial cells. Overexpression of eNOS and HA-tagged PAI-1 in COS7 cells confirmed the colocalization observations in HUVECs. Furthermore, the source of intracellular PAI-1 interacting with eNOS was shown to be endocytosis derived. The interaction between PAI-1 and eNOS is a direct interaction as supported in experiments with purified proteins. Moreover, PAI-1 directly inhibits eNOS activity, reducing NO synthesis, and the knockdown or antagonism of PAI-1 increases NO bioavailability. Taken together, these findings place PAI-1 as a negative regulator of eNOS and disruptions in eNOS-PAI-1 binding promote increases in NO production and enhance vasodilation in vivo.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Nitric Oxide Synthase Type III/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Biological Availability , Cell Line , Gene Expression Regulation, Enzymologic/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Nitric Oxide , Nitric Oxide Synthase Type III/genetics , Piperazines/pharmacology , Plasminogen Activator Inhibitor 1/genetics , Protein Binding , Vasodilation/drug effects , Vasodilation/physiology , para-Aminobenzoates/pharmacology
10.
J Biol Chem ; 296: 100336, 2021.
Article in English | MEDLINE | ID: mdl-33508317

ABSTRACT

Soluble guanylate cyclase (sGC) is a heme-containing heterodimeric enzyme that generates many molecules of cGMP in response to its ligand nitric oxide (NO); sGC thereby acts as an amplifier in NO-driven biological signaling cascades. Because sGC helps regulate the cardiovascular, neuronal, and gastrointestinal systems through its cGMP production, boosting sGC activity and preventing or reversing sGC inactivation are important therapeutic and pharmacologic goals. Work over the last two decades is uncovering the processes by which sGC matures to become functional, how sGC is inactivated, and how sGC is rescued from damage. A diverse group of small molecules and proteins have been implicated in these processes, including NO itself, reactive oxygen species, cellular heme, cell chaperone Hsp90, and various redox enzymes as well as pharmacologic sGC agonists. This review highlights their participation and provides an update on the processes that enable sGC maturation, drive its inactivation, or assist in its recovery in various settings within the cell, in hopes of reaching a better understanding of how sGC function is regulated in health and disease.


Subject(s)
Soluble Guanylyl Cyclase/metabolism , Animals , Dimerization , Glyceraldehyde-3-Phosphate Dehydrogenases , HSP90 Heat-Shock Proteins/metabolism , Heme/metabolism , Humans , Ligands , Nitric Oxide/metabolism , Oxidation-Reduction , Soluble Guanylyl Cyclase/antagonists & inhibitors
11.
Biol Chem ; 403(11-12): 1005-1015, 2022 11 25.
Article in English | MEDLINE | ID: mdl-36152339

ABSTRACT

The intracellular trafficking of mitochondrial heme presents a fundamental challenge to animal cells. This article provides some background on heme allocation, discusses some of the concepts, and then reviews research done over the last decade, much in the author's laboratory, that is uncovering unexpected and important roles for glyceraldehyde 3-phosphate dehydrogenase (GAPDH), heat shock protein 90 (hsp90), and nitric oxide (NO) in enabling and regulating the allocation of mitochondrial heme to hemeproteins that mature and function outside of the mitochondria. A model for how hemeprotein functions can be regulated in cells through the coordinate participation of GAPDH, hsp90, and NO in allocating cellular heme is presented.


Subject(s)
Heme , Hemeproteins , Animals , Heme/metabolism , Nitric Oxide/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , HSP90 Heat-Shock Proteins/metabolism , Mammals/metabolism
12.
Biol Chem ; 403(11-12): 1043-1053, 2022 Nov 25.
Article in English | MEDLINE | ID: mdl-36302634

ABSTRACT

Heme regulatory motifs (HRMs) are found in a variety of proteins with diverse biological functions. In heme oxygenase-2 (HO2), heme binds to the HRMs and is readily transferred to the catalytic site in the core of the protein. To further define this heme transfer mechanism, we evaluated the ability of GAPDH, a known heme chaperone, to transfer heme to the HRMs and/or the catalytic core of HO2. Our results indicate GAPDH and HO2 form a complex in vitro. We have followed heme insertion at both sites by fluorescence quenching in HEK293 cells with HO2 reporter constructs. Upon mutation of residues essential for heme binding at each site in our reporter construct, we found that HO2 binds heme at the core and the HRMs in live cells and that heme delivery to HO2 is dependent on the presence of GAPDH that is competent for heme binding. In sum, GAPDH is involved in heme delivery to HO2 but, surprisingly, not to a specific site on HO2. Our results thus emphasize the importance of heme binding to both the core and the HRMs and the interplay of HO2 with the heme pool via GAPDH to maintain cellular heme homeostasis.


Subject(s)
Heme Oxygenase (Decyclizing) , Heme , Humans , Heme/chemistry , HEK293 Cells , Heme Oxygenase (Decyclizing)/chemistry , Heme Oxygenase (Decyclizing)/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/chemistry , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism
13.
Biophys J ; 120(23): 5196-5206, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34748763

ABSTRACT

Mechanisms that regulate nitric oxide synthase enzymes (NOS) are of interest in biology and medicine. Although NOS catalysis relies on domain motions and is activated by calmodulin (CaM) binding, the relationships are unclear. We used single-molecule fluorescence resonance energy transfer (FRET) spectroscopy to elucidate the conformational states distribution and associated conformational fluctuation dynamics of the two NOS electron transfer domains in an FRET dye-labeled endothelial NOS reductase domain (eNOSr) and to understand how CaM affects the dynamics to regulate catalysis by shaping the spatial and temporal conformational behaviors of eNOSr. In addition, we developed and applied a new imaging approach capable of recording three-dimensional FRET efficiency versus time images to characterize the impact on dynamic conformal states of the eNOSr enzyme by the binding of CaM, which identifies clearly that CaM binding generates an extra new open state of eNOSr, resolving more detailed NOS conformational states and their fluctuation dynamics. We identified a new output state that has an extra open conformation that is only populated in the CaM-bound eNOSr. This may reveal the critical role of CaM in triggering NOS activity as it gives conformational flexibility for eNOSr to assume the electron transfer output FMN-heme state. Our results provide a dynamic link to recently reported EM static structure analyses and demonstrate a capable approach in probing and simultaneously analyzing all of the conformational states, their fluctuations, and the fluctuation dynamics for understanding the mechanism of NOS electron transfer, involving electron transfer among FAD, FMN, and heme domains, during nitric oxide synthesis.


Subject(s)
Calmodulin , Nitric Oxide Synthase Type III , Calmodulin/metabolism , Electron Transport , Heme/metabolism , Nitric Oxide , Nitric Oxide Synthase , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type III/metabolism
14.
J Biol Chem ; 295(24): 8145-8154, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32358060

ABSTRACT

Soluble guanylyl cyclase (sGC) is a key component of NO-cGMP signaling in mammals. Although heme must bind in the sGC ß1 subunit (sGCß) for sGC to function, how heme is delivered to sGCß remains unknown. Given that GAPDH displays properties of a heme chaperone for inducible NO synthase, here we investigated whether heme delivery to apo-sGCß involves GAPDH. We utilized an sGCß reporter construct, tetra-Cys sGCß, whose heme insertion can be followed by fluorescence quenching in live cells, assessed how lowering cell GAPDH expression impacts heme delivery, and examined whether expressing WT GAPDH or a GAPDH variant defective in heme binding recovers heme delivery. We also studied interaction between GAPDH and sGCß in cells and their complex formation and potential heme transfer using purified proteins. We found that heme delivery to apo-sGCß correlates with cellular GAPDH expression levels and depends on the ability of GAPDH to bind intracellular heme, that apo-sGCß associates with GAPDH in cells and dissociates when heme binds sGCß, and that the purified GAPDH-heme complex binds to apo-sGCß and transfers its heme to sGCß. On the basis of these results, we propose a model where GAPDH obtains mitochondrial heme and then forms a complex with apo-sGCß to accomplish heme delivery to sGCß. Our findings illuminate a critical step in sGC maturation and uncover an additional mechanism that regulates its activity in health and disease.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Heme/metabolism , Soluble Guanylyl Cyclase/metabolism , Animals , Apoproteins/metabolism , HEK293 Cells , Heme/pharmacology , Humans , Kinetics , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Protein Binding/drug effects , Protein Multimerization/drug effects , Rats
15.
J Biol Chem ; 295(8): 2203-2211, 2020 02 21.
Article in English | MEDLINE | ID: mdl-31914408

ABSTRACT

Production of reactive oxygen species caused by dysregulated endothelial nitric-oxide synthase (eNOS) activity is linked to vascular dysfunction. eNOS is a major target protein of the primary calcium-sensing protein calmodulin. Calmodulin is often modified by the main biomarker of nitroxidative stress, 3-nitrotyrosine (nitroTyr). Despite nitroTyr being an abundant post-translational modification on calmodulin, the mechanistic role of this modification in altering calmodulin function and eNOS activation has not been investigated. Here, using genetic code expansion to site-specifically nitrate calmodulin at its two tyrosine residues, we assessed the effects of these alterations on calcium binding by calmodulin and on binding and activation of eNOS. We found that nitroTyr-calmodulin retains affinity for eNOS under resting physiological calcium concentrations. Results from in vitro eNOS assays with calmodulin nitrated at Tyr-99 revealed that this nitration reduces nitric-oxide production and increases eNOS decoupling compared with WT calmodulin. In contrast, calmodulin nitrated at Tyr-138 produced more nitric oxide and did so more efficiently than WT calmodulin. These results indicate that the nitroTyr post-translational modification, like tyrosine phosphorylation, can impact calmodulin sensitivity for calcium and reveal Tyr site-specific gain or loss of functions for calmodulin-induced eNOS activation.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Nitric Oxide Synthase Type III/metabolism , Tyrosine/metabolism , Animals , Cattle , Cell Extracts , Fluorescence , HEK293 Cells , Humans , Interferometry , Nitrosation , Protein Binding
16.
Am J Physiol Lung Cell Mol Physiol ; 320(5): L739-L749, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33596129

ABSTRACT

Idiopathic pulmonary arterial hypertension (IPAH) is a rapidly progressive disease with several treatment options. Long-term mortality remains high with great heterogeneity in treatment response. Even though most of the pathology of IPAH is observed in the lung, there is systemic involvement. Platelets from patients with IPAH have characteristic metabolic shifts and defects in activation; therefore, we investigated whether they could be used to identify other disease-specific abnormalities. We used proteomics to investigate protein expression changes in platelets from patients with IPAH compared with healthy controls. Key abnormalities of nitric oxide pathway were tested in platelets from a larger cohort of unique patients with IPAH. Platelets showed abnormalities in the prostacyclin and nitric oxide pathways, which are dysregulated in IPAH and hence targets of therapy. We detected reduced expression of G protein αs and increased expression of the regulatory subunits of the cAMP-dependent protein kinase (PKA) type II isoforms, supporting an overall decrease in the activation of the prostacyclin pathway. We noted reduced levels of the soluble guanylate cyclase (sGC) subunits and increased expression of the phosphodiesterase type 5 A (PDE5A), conditions that affect the response to nitric oxide. Ensuing analysis of 38 unique patients with IPAH demonstrated considerable variation in the levels and specific activity of sGC, a finding with novel implications for personalized therapy. Platelets have some of the characteristic vasoactive signal abnormalities seen in IPAH and may provide comprehensive ex vivo mechanistic information to direct therapeutic decisions.


Subject(s)
Blood Platelets/pathology , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Familial Primary Pulmonary Hypertension/physiopathology , Proteome/metabolism , Soluble Guanylyl Cyclase/metabolism , Adult , Aged , Blood Platelets/metabolism , Case-Control Studies , Female , Humans , Male , Middle Aged , Proteome/analysis , Young Adult
17.
Proc Natl Acad Sci U S A ; 115(6): E1117-E1126, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29358373

ABSTRACT

Maturation of adult (α2ß2) and fetal hemoglobin (α2γ2) tetramers requires that heme be incorporated into each globin. While hemoglobin alpha (Hb-α) relies on a specific erythroid chaperone (alpha Hb-stabilizing protein, AHSP), the other chaperones that may help mature the partner globins (Hb-γ or Hb-ß) in erythroid cells, or may enable nonerythroid cells to express mature Hb, are unknown. We investigated a role for heat-shock protein 90 (hsp90) in Hb maturation in erythroid precursor cells that naturally express Hb-α with either Hb-γ (K562 and HiDEP-1 cells) or Hb-ß (HUDEP-2) and in nonerythroid cell lines that either endogenously express Hb-αß (RAW and A549) or that we transfected to express the globins. We found the following: (i) AHSP and hsp90 associate with distinct globin partners in their immature heme-free states (AHSP with apo-Hbα, and hsp90 with apo-Hbß or Hb-γ) and that hsp90 does not associate with mature Hb. (ii) Hsp90 stabilizes the apo-globins and helps to drive their heme insertion reactions, as judged by pharmacologic hsp90 inhibition or by coexpression of an ATP-ase defective hsp90. (iii) In nonerythroid cells, heme insertion into all globins became hsp90-dependent, which may explain how mixed Hb tetramers can mature in cells that do not express AHSP. Together, our findings uncover a process in which hsp90 first binds to immature, heme-free Hb-γ or Hb-ß, drives their heme insertion process, and then dissociates to allow their heterotetramer formation with Hb-α. Thus, in driving heme insertion, hsp90 works in concert with AHSP to generate functional Hb tetramers during erythropoiesis.


Subject(s)
Erythroid Precursor Cells/metabolism , Erythropoiesis/physiology , HSP90 Heat-Shock Proteins/metabolism , Heme/metabolism , Hemoglobins/biosynthesis , Lung/metabolism , Macrophages/metabolism , Blood Proteins/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Erythroid Precursor Cells/cytology , Heme/chemistry , Hemoglobins/chemistry , Humans , Lung/cytology , Macrophages/cytology , Molecular Chaperones/metabolism , Protein Binding
18.
Am J Respir Cell Mol Biol ; 62(1): 43-48, 2020 01.
Article in English | MEDLINE | ID: mdl-31340135

ABSTRACT

The soluble guanylyl cyclase (sGC)-cyclic guanosine monophosphate signaling pathway evokes vascular smooth muscle relaxation; whether this pathway mediates airway smooth muscle relaxation remains controversial. We posit that sGC activators are equi-effective as ß-agonists in reversing contractile agonist-induced airway smooth muscle shortening. To provide clarity, we tested the efficacy of sGC stimulator and activator drugs, BAY 41-2272 and BAY 60-2270, respectively, in reversing bronchoconstriction of human small airways using human precision-cut lung slices (hPCLS). Both BAY drugs reversed carbachol-induced bronchoconstriction to a maximal degree comparable to that of formoterol. Moreover, the sGC drugs remained effective bronchodilators despite formoterol-induced desensitization of the airways. Analysis of the hPCLS after their activation by sGC or ß2-adrenergic receptor agonist showed distinct cyclic nucleotide accumulation in the hPCLS. Collectively, these data suggest that cAMP and cyclic guanosine monophosphate pathways are equi-effective for reversing carbachol-induced bronchoconstriction in the human airway via separate and distinct second messenger pathways. This should open the door for future studies to test whether sGC-targeted drugs alone or in combination can serve as effective bronchodilators in asthma and chronic obstructive pulmonary disease.


Subject(s)
Bronchodilator Agents/pharmacology , Muscle, Smooth/drug effects , Respiratory System/drug effects , Soluble Guanylyl Cyclase/metabolism , Asthma/drug therapy , Asthma/metabolism , Bronchoconstriction/drug effects , Cyclic GMP/metabolism , Humans , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Muscle, Smooth/metabolism , Nitric Oxide/metabolism , Respiratory System/metabolism , Signal Transduction/drug effects , Trachea/drug effects , Trachea/metabolism
19.
J Biol Chem ; 294(19): 7904-7916, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30926606

ABSTRACT

Nitric oxide (NO) synthases (NOSs) catalyze the formation of NO from l-arginine. We have shown previously that the NOS enzyme catalytic cycle involves a large number of reactions but can be characterized by a global model with three main rate-limiting steps. These are the rate of heme reduction by the flavin domain (kr ), of dissociation of NO from the ferric heme-NO complex (kd ), and of oxidation of the ferrous heme-NO complex (kox). The reaction of oxygen with the ferrous heme-NO species is part of a futile cycle that does not directly contribute to NO synthesis but allows a population of inactive enzyme molecules to return to the catalytic cycle, and thus, enables a steady-state NO synthesis rate. Previously, we have reported that this reaction does involve the reaction of oxygen with the NO-bound ferrous heme complex, but the mechanistic details of the reaction, that could proceed via either an inner-sphere or an outer-sphere mechanism, remained unclear. Here, we present additional experiments with neuronal NOS (nNOS) and inducible NOS (iNOS) variants (nNOS W409F and iNOS K82A and V346I) and computational methods to study how changes in heme access and electronics affect the reaction. Our results support an inner-sphere mechanism and indicate that the particular heme-thiolate environment of the NOS enzymes can stabilize an N-bound FeIII-N(O)OO- intermediate species and thereby catalyze this reaction, which otherwise is not observed or favorable in proteins like globins that contain a histidine-coordinated heme.


Subject(s)
Models, Chemical , Nitric Oxide Synthase Type II/chemistry , Nitric Oxide Synthase Type I/chemistry , Nitric Oxide/chemistry , Amino Acid Substitution , Animals , Heme , Mice , Mutation, Missense , Nitric Oxide/genetics , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oxidation-Reduction , Rats
20.
J Biol Chem ; 294(35): 12880-12891, 2019 08 30.
Article in English | MEDLINE | ID: mdl-31311859

ABSTRACT

The enzyme soluble guanylyl cyclase (sGC) is a heterodimer composed of an α subunit and a heme-containing ß subunit. It participates in signaling by generating cGMP in response to nitric oxide (NO). Heme insertion into the ß1 subunit of sGC (sGCß) is critical for function, and heat shock protein 90 (HSP90) associates with heme-free sGCß (apo-sGCß) to drive its heme insertion. Here, we tested the accuracy and relevance of a modeled apo-sGCß-HSP90 complex by constructing sGCß variants predicted to have an impaired interaction with HSP90. Using site-directed mutagenesis, purified recombinant proteins, mammalian cell expression, and fluorescence approaches, we found that (i) three regions in apo-sGCß predicted by the model mediate direct complex formation with HSP90 both in vitro and in mammalian cells; (ii) such HSP90 complex formation directly correlates with the extent of heme insertion into apo-sGCß and with cyclase activity; and (iii) apo-sGCß mutants possessing an HSP90-binding defect instead bind to sGCα in cells and form inactive, heme-free sGC heterodimers. Our findings uncover the molecular features of the cellular apo-sGCß-HSP90 complex and reveal its dual importance in enabling heme insertion while preventing inactive heterodimer formation during sGC maturation.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Soluble Guanylyl Cyclase/metabolism , Animals , COS Cells , Cattle , Cells, Cultured , Chlorocebus aethiops , Humans
SELECTION OF CITATIONS
SEARCH DETAIL