Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Sci Rep ; 14(1): 14273, 2024 06 20.
Article in English | MEDLINE | ID: mdl-38902362

ABSTRACT

Tumor-derived extracellular vesicles (EVs) show great potential as biomarkers for several diseases, including pancreatic cancer, due to their roles in cancer development and progression. However, the challenge of utilizing EVs as biomarkers lies in their inherent heterogeneity in terms of size and concentration, making accurate quantification difficult, which is highly dependent on the isolation and quantification methods used. In our study, we compared three EV isolation techniques and two EV quantification methods. We observed variations in EV concentration, with approximately 1.5-fold differences depending on the quantification method used. Interestingly, all EV isolation techniques consistently yielded similar EV quantities, overall size distribution, and modal sizes. In contrast, we found a notable increase in total EV amounts in samples from pancreatic cancer cell lines, mouse models, and patient plasma, compared to non-cancerous conditions. Moreover, individual tumor-derived EVs exhibited at least a 3-fold increase in several EV biomarkers. Our data, obtained from EVs isolated using various techniques and quantified through different methods, as well as originating from various pancreatic cancer models, suggests that EV profiling holds promise for the identification of unique and cancer-specific biomarkers in pancreatic cancer.


Subject(s)
Biomarkers, Tumor , Epithelial Cell Adhesion Molecule , Extracellular Vesicles , Glypicans , Pancreatic Neoplasms , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Extracellular Vesicles/metabolism , Humans , Biomarkers, Tumor/metabolism , Animals , Mice , Cell Line, Tumor , Epithelial Cell Adhesion Molecule/metabolism , Glypicans/metabolism , Integrin alphaV/metabolism
2.
3.
Clin Sci (Lond) ; 137(11): 913-930, 2023 06 14.
Article in English | MEDLINE | ID: mdl-37254732

ABSTRACT

Compromised barrier function of colon epithelium with aging is largely due to gut microbial dysbiosis. Recent studies implicate an important role for angiotensin converting enzymes, ACE and ACE2, angiotensins, and the receptors, AT1 receptor (AT1R) and Mas receptor (MasR), in the regulation of colon functions. The present study tested the hypothesis that leaky gut in aging is associated with an imbalance in ACE2/ACE and that the treatment with angiotenisn-(1-7) (Ang-(1-7)) will restore gut barrier integrity and microbiome. Studies were carried out in Young (3-4 months) and old (20-24 months) male mice. Ang-(1-7) was administered by using osmotic pumps. Outcome measures included expressions of ACE, ACE2, AT1R, and MasR, intestinal permeability by using FITC-dextran, and immunohistochemistry of claudin 1 and occludin, and intestinal stem cells (ISCs). ACE2 protein and activity were decreased in Old group while that of ACE were unchanged. Increased intestinal permeability and plasma levels of zonulin-1 in the Old group were normalized by Ang-(1-7). Epithelial disintegrity, reduced number of goblet cells and ISCs in the old group were restored by Ang-(1-7). Expression of claudin 1 and occludin in the aging colon was increased by Ang-(1-7). Infiltration of CD11b+ or F4/80+ inflammatory cells in the old colons were decreased by Ang-(1-7). Gut microbial dysbiosis in aging was evident by decreased richness and altered beta diversity that were reversed by Ang-(1-7) with increased abundance of Lactobacillus or Lachnospiraceae. The present study shows that Ang-(1-7) restores gut barrier integrity and reduces inflammation in the aging colon by restoring the layer of ISCs and by restructuring the gut microbiome.


Subject(s)
Gastrointestinal Microbiome , Mice , Male , Animals , Angiotensin-Converting Enzyme 2 , Dysbiosis , Claudin-1 , Occludin , Angiotensin I/pharmacology , Angiotensin I/metabolism , Peptidyl-Dipeptidase A/metabolism , Peptide Fragments/pharmacology , Peptide Fragments/metabolism , Aging , Angiotensin II/metabolism
4.
Sci Rep ; 13(1): 2543, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36782016

ABSTRACT

Aging is associated with chronic systemic inflammation largely due to increased myelopoiesis, which in turn increases risk for vascular disease. We have previously shown evidence for the therapeutic potential of Angiotensin-(1-7) (Ang-(1-7)) in reversing vasoreparative dysfunction in aging. This study tested the hypothesis that ischemic vascular repair in aging by Ang-(1-7) involves attenuation of myelopoietic potential in the bone marrow and decreased mobilization of inflammatory cells. Young or Old male mice of age 3-4 and 22-24 months, respectively, received Ang-(1-7) (1 µg/kg/min, s.c.) for four weeks. Myelopoiesis was evaluated in the bone marrow (BM) cells by carrying out the colony forming unit (CFU-GM) assay followed by flow cytometry of monocyte-macrophages. Expression of pro-myelopoietic factors and alarmins in the hematopoietic progenitor-enriched BM cells was evaluated. Hindlimb ischemia (HLI) was induced by femoral ligation, and mobilization of monocytes into the blood stream was determined. Blood flow recovery was monitored by Laser Doppler imaging and infiltration of inflammatory cells was evaluated by immunohistochemistry. BM cells from Old mice generated a higher number of monocytes (Ly6G-CD11b+Ly6Chi) and M1 macrophages (Ly6ChiF4/80+) compared to that of Young, which was reversed by Ang-(1-7). Gene expression of selected myelopoietic factors, alarmins (S100A8, S100A9, S100A14 and HMGb1) and the receptor for alarmins, RAGE, was higher in the Old hematopoietic progenitor-enriched BM cells compared to the Young. Increased expressions of these factors were decreased by Ang-(1-7). Ischemia-induced mobilization of monocytes was higher in Old mice with decreased blood flow recovery and increased infiltration of monocyte-macrophages compared to the Young, all of which were reversed by Ang-(1-7). Enhanced ischemic vascular repair by Ang-(1-7) in aging is largely by decreasing the generation and recruitment of inflammatory monocyte-macrophages to the areas of ischemic injury. This is associated with decreased alarmin signaling in the BM-hematopoietic progenitor cells.


Subject(s)
Alarmins , Myelopoiesis , Mice , Male , Animals , Hematopoietic Stem Cells , Ischemia , Inflammation
5.
Mol Pharmacol ; 99(1): 29-38, 2021 01.
Article in English | MEDLINE | ID: mdl-32321734

ABSTRACT

Bone marrow-derived hematopoietic stem/progenitor cells are vasculogenic and play an important role in endothelial health and vascular homeostasis by participating in postnatal vasculogenesis. Progenitor cells are mobilized from bone marrow niches in response to remote ischemic injury and migrate to the areas of damage and stimulate revascularization largely by paracrine activation of angiogenic functions in the peri-ischemic vasculature. This innate vasoprotective mechanism is impaired in certain chronic clinical conditions, which leads to the development of cardiovascular complications. Members of the renin-angiotensin system-angiotensin-converting enzymes (ACEs) ACE and ACE2, angiotensin II (Ang II), Ang-(1-7), and receptors AT1 and Mas-are expressed in vasculogenic progenitor cells derived from humans and rodents. Ang-(1-7), generated by ACE2, is known to produce cardiovascular protective effects by acting on Mas receptor and is considered as a counter-regulatory mechanism to the detrimental effects of Ang II. Evidence has now been accumulating in support of the activation of the ACE2/Ang-(1-7)/Mas receptor pathway by pharmacologic or molecular maneuvers, which stimulates mobilization of progenitor cells from bone marrow, migration to areas of vascular damage, and revascularization of ischemic areas in pathologic conditions. This minireview summarizes recent studies that have enhanced our understanding of the physiology and pharmacology of vasoprotective axis in bone marrow-derived progenitor cells in health and disease. SIGNIFICANCE STATEMENT: Hematopoietic stem progenitor cells (HSPCs) stimulate revascularization of ischemic areas. However, the reparative potential is diminished in certain chronic clinical conditions, leading to the development of cardiovascular diseases. ACE2 and Mas receptor are key members of the alternative axis of the renin-angiotensin system and are expressed in HSPCs. Accumulating evidence points to activation of ACE2 or Mas receptor as a promising approach for restoring the reparative potential, thereby preventing the development of ischemic vascular diseases.


Subject(s)
Angiotensin I/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Cardiovascular Diseases/metabolism , Drug Delivery Systems/trends , Hematopoietic Stem Cells/metabolism , Peptide Fragments/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cardiovascular Agents/administration & dosage , Cardiovascular Agents/metabolism , Cardiovascular Diseases/drug therapy , Drug Delivery Systems/methods , Hematopoietic Stem Cells/drug effects , Humans , Proto-Oncogene Mas , Signal Transduction/drug effects , Signal Transduction/physiology , Stem Cells/drug effects , Stem Cells/metabolism
6.
Br J Pharmacol ; 176(22): 4373-4387, 2019 11.
Article in English | MEDLINE | ID: mdl-30367728

ABSTRACT

BACKGROUND AND PURPOSE: CD34+ haematopoietic stem/progenitor cells have revascularization potential and are now being tested for the treatment of ischaemic vascular diseases in clinical trials. We tested the hypothesis that mitochondrial depolarization stimulates the reparative functions of CD34+ cells. EXPERIMENTAL APPROACH: Peripheral blood was obtained from healthy individuals (n = 63), and mononuclear cells (MNCs) were separated. MNCs were enriched for lineage negative cells, followed by isolation of CD34+ cells. Vascular repair-relevant functions of CD34+ cells, proliferation and migration, were evaluated in the presence and absence of diazoxide. Mitochondrial membrane potential, ROS and NO levels were evaluated by flow cytometry by using JC-1, mitoSOX and DAF-FM respectively. KEY RESULTS: Diazoxide stimulated the proliferation and migration of CD34+ cells that were comparable to the responses induced by stromal-derived factor-1α (SDF) or VEGF. Effects of diazoxide were blocked by either 5-hydroxydecanoate (5HD), a selective mitochondrial ATP-sensitive potassium channel (mitoKATP ) inhibitor, or by L-NAME. Diazoxide induced mitochondrial depolarization, and NO and cGMP generation that were 5HD-sensitive. The generation of NO and cGMP by diazoxide was blocked by an endothelial NOS (eNOS)-selective inhibitor, NIO, but not by a neuronal (n)NOS-selective inhibitor, Nω -propyl-L-arginine (NPA). A Ca2+ chelator, BAPTA, Akt inhibitor, triciribine, or PI3K inhibitor, LY294002, inhibited the NO release induced by diazoxide. Phosphorylation of eNOS at Ser1177 and dephosphorylation at Thr495 were increased. Diazoxide-induced ROS generation and phosphorylation of eNOS at Ser1177 were reduced by NPA. CONCLUSION AND IMPLICATIONS: Diazoxide stimulates vascular repair-relevant functions of CD34+ cells via the mitoKATP -dependent release of NO and ROS. LINKED ARTICLES: This article is part of a themed section on Mitochondrial Pharmacology: Featured Mechanisms and Approaches for Therapy Translation. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.22/issuetoc.


Subject(s)
Antigens, CD34 , Diazoxide/pharmacology , Hematopoietic Stem Cells/drug effects , Mitochondria/physiology , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cyclic GMP/metabolism , Hematopoietic Stem Cells/physiology , Humans
7.
Methods Mol Biol ; 1614: 47-59, 2017.
Article in English | MEDLINE | ID: mdl-28500594

ABSTRACT

In recent years, previously unknown functions have been conferred to the RAAS and have been explored in mechanistic studies and disease models. Implication of bone marrow stem/progenitor cells in the cardiovascular protective or detrimental effects of RAAS is a prominent advancement because of the translational significance. Selected members of RAAS are now known to modulate migration, proliferation, and mobilization of bone marrow cells in response to ischemic insult, which are sensitive indicators of vascular repair-relevant functions. In this Chapter, protocols for most frequently used, in vitro, ex vivo, and in vivo assays to explore the potential of RAAS members to stimulate vascular repair-relevant functions of bone marrow stem/progenitor cells of human and murine origin.


Subject(s)
Bone Marrow Cells/metabolism , Cell Migration Assays/methods , Immunomagnetic Separation/methods , Renin-Angiotensin System/physiology , Stem Cells/metabolism , Animals , Cell Proliferation , Cells, Cultured , Colony-Forming Units Assay/methods , Flow Cytometry/methods , Humans , Mice , Mice, Inbred C57BL
8.
Diabetes ; 66(2): 505-518, 2017 02.
Article in English | MEDLINE | ID: mdl-27856608

ABSTRACT

The angiotensin (ANG)-(1-7)/Mas receptor (MasR) pathway activates vascular repair-relevant functions of bone marrow progenitor cells. We tested the effects of ANG-(1-7) on mobilization and vasoreparative functions of progenitor cells that are impaired in diabetes. The study was performed in streptozotocin-induced diabetic (db/db) mice. Diabetes resulted in a decreased number of Lineage-Sca-1+c-Kit+ (LSK) cells in the circulation, which was normalized by ANG-(1-7). Diabetes-induced depletion of LSK cells in the bone marrow was reversed by ANG-(1-7). ρ-Kinase (ROCK) activity was increased specifically in bone marrow LSK cells by ANG-(1-7) in diabetes, and the beneficial effects of ANG-(1-7) were prevented by fasudil. ANG-(1-7) increased Slit3 levels in the bone marrow supernatants, which activated ROCK in LSK cells and sensitized them for stromal-derived factor-1α (SDF)-induced migration. Diabetes prevented the mobilization of LSK cells in response to ischemia and impaired the recovery of blood flow, both of which were reversed by ANG-(1-7) in both models of diabetes. Genetic ablation of MasR prevented ischemia-induced mobilization of LSK cells and impaired blood flow recovery, which was associated with decreased proliferation and migration of LSK cells in response to SDF or vascular endothelial growth factor. These results suggest that MasR is a promising target for the treatment of diabetic bone marrow mobilopathy and vascular disease.


Subject(s)
Angiotensin I/pharmacology , Blood Vessels/drug effects , Bone Marrow Cells/drug effects , Cell Movement/drug effects , Diabetes Mellitus, Experimental/physiopathology , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Stem Cells/drug effects , Vasodilator Agents/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Blood Vessels/physiopathology , Bone Marrow/drug effects , Bone Marrow/physiopathology , Cell Lineage , Chemokine CXCL12/drug effects , Chemokine CXCL12/metabolism , Ischemia , Male , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Mice , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Recovery of Function/drug effects , Regeneration , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/metabolism , rho-Associated Kinases/drug effects , rho-Associated Kinases/metabolism
9.
Sci Rep ; 6: 26131, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27188595

ABSTRACT

Diabetes is associated with impaired mobilization of bone marrow stem/progenitor cells that accelerate vascularization of ischemic areas. This study characterized mobilization of vascular reparative bone marrow progenitor cells in mouse models of diabetes. Age-matched control or streptozotocin (STZ)-induced diabetic, and db/db mice with lean-controls were studied. Mobilization induced by G-CSF, AMD3100 or ischemia was evaluated by flow cytometric enumeration of circulating Lin(-)Sca-1(+)cKit(+) (LSK) cells, and by colony forming unit (CFU) assay. The circulating WBCs and LSKs, and CFUs were reduced in both models with a shorter duration (10-12 weeks) of diabetes compared to their respective controls. Longer duration of STZ-diabetes (≥20 weeks) induced impairment of G-CSF- or AMD3100-mobilization (P < 0.01, n = 8). In db/db mice, mobilization by G-CSF or AMD3100 was either increased or unaffected (P < 0.05, n = 6 to 8). Proliferation, migration, and ischemia-induced mobilization, of LSK cells were impaired in both models. Leptin receptor antagonist, PESLAN-1, increased G-CSF- or AMD3100-mobilization of WBCs and LSKs, compared to the untreated. Leptin increased basal WBCs, decreased basal and AMD3100-mobilized LSK cells, and had no effect on G-CSF. These results suggest that mobilopathy is apparent in STZ-diabetes but not in db/db mice. Leptin receptor antagonism would be a promising approach for reversing diabetic bone marrow mobilopathy.


Subject(s)
Bone Marrow Cells/physiology , Diabetes Mellitus, Experimental/pathology , Neovascularization, Physiologic , Receptors, Leptin/deficiency , Stem Cells/physiology , Animals , Cell Movement , Cell Proliferation , Colony-Forming Units Assay , Flow Cytometry , Mice
10.
Am J Physiol Heart Circ Physiol ; 309(10): H1697-707, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26386115

ABSTRACT

CD34(+) stem/progenitor cells have been identified as a promising cell population for the autologous cell-based therapies in patients with cardiovascular disease. The counter-regulatory axes of renin angiotensin system, angiotensin converting enzyme (ACE)/Ang II/angiotensin type 1 (AT1) receptor and ACE2/Ang-(1-7)/Mas receptor, play an important role in the cardiovascular repair. This study evaluated the expression and vascular repair-relevant functions of these two pathways in human CD34(+) cells. CD34(+) cells were isolated from peripheral blood mononuclear cells (MNCs), obtained from healthy volunteers. Expression of ACE, ACE2, AT1, and angiotensin type 2 and Mas receptors were determined. Effects of Ang II, Ang-(1-7), Norleu(3)-Ang-(1-7), and ACE2 activators, xanthenone (XNT) and diminazene aceturate (DIZE) on proliferation, migration, and adhesion of CD34(+) cells were evaluated. ACE2 and Mas were relatively highly expressed in CD34(+) cells compared with MNCs. Ang-(1-7) or its analog, Norleu(3)-Ang-(1-7), stimulated proliferation of CD34(+) cells that was associated with decrease in phosphatase and tensin homologue deleted on chromosome 10 levels and was inhibited by triciribin, an AKT inhibitor. Migration of CD34(+) cells was enhanced by Ang-(1-7) or Norleu(3)-Ang-(1-7) that was decreased by a Rho-kinase inhibitor, Y-27632. In the presence of Ang II, XNT or DIZE enhanced proliferation and migration that were blocked by DX-600, an ACE2 inhibitor. Treatment of MNCs with Ang II, before the isolation of CD34(+) cells, attenuated the proliferation and migration to stromal derived factor-1α. This attenuation was reversed by apocynin, an NADPH oxidase inhibitor. Adhesion of MNCs or CD34(+) cells to fibronectin was enhanced by Ang II and was unaffected by Ang-(1-7). This study suggests that ACE2/Ang-(1-7)/Mas pathway stimulates functions of CD34(+) cells that are cardiovascular protective, whereas Ang II attenuates these functions by acting on MNCs. These findings imply that activation of ACE2/Ang-(1-7)/Mas axis is a promising approach for enhancing reparative outcomes of cell-based therapies.


Subject(s)
Angiotensin II/pharmacology , Angiotensin I/pharmacology , Leukocytes, Mononuclear/drug effects , Peptide Fragments/pharmacology , Peptidyl-Dipeptidase A/drug effects , Proto-Oncogene Proteins/drug effects , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 2/drug effects , Receptors, G-Protein-Coupled/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology , Angiotensin-Converting Enzyme 2 , Antigens, CD34 , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Diminazene/analogs & derivatives , Diminazene/pharmacology , Humans , Leukocytes, Mononuclear/metabolism , Peptidyl-Dipeptidase A/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Receptors, G-Protein-Coupled/metabolism , Wound Healing/drug effects , Xanthones/pharmacology
11.
J Sex Med ; 11(9): 2153-63, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24953642

ABSTRACT

INTRODUCTION: Angiotensin (Ang)-(1-7) is a recently identified vasoprotective heptapeptide, and it appears to activate the reparative functions of bone marrow-derived stem/progenitor cells (BMPCs). AIM: This study evaluated the effect of Ang-(1-7) in the angiogenic function of cavernosum in type 1 diabetes (T1D) and delineated the role of BMPCs in this protective function. METHODS: T1D was induced by streptozotocin in mice, and mice with 20-24 weeks of diabetes were used for the study. Ang-(1-7) was administered subcutaneously by using osmotic pumps. Cavernosa, and BMPCs from peripheral blood and bone marrow were evaluated in different assay systems. MAIN OUTCOME MEASURES: Angiogenic function was determined by endothelial tube formation in matrigel assay. Circulating BMPCs were enumerated by flow cytometry and proliferation was determined by BrdU incorporation. Cell-free supernatant of BMPCs were collected and tested for paracrine angiogenic effect. Expression of angiogenic factors in BMPCs and cavernosa were determined by real-time polymerase chain reaction. RESULTS: Ang-(1-7) (100 nM) stimulated angiogenesis in mouse cavernosum that was partially inhibited by Mas1 receptor antagonist, A779 (10 µM) (P < 0.05). In cavernosa of T1D, the angiogenic responses to Ang-(1-7) (P < 0.005) and VEGF (100 nM) (P < 0.03) were diminished. Ang-(1-7) treatment for 4 weeks reversed T1D-induced decrease in the VEGF-mediated angiogenesis. Ang-(1-7) treatment increased the circulating number of BMPCs and proliferation that were decreased in T1D (P < 0.02). Paracrine angiogenic function of BMPCs was reduced in diabetic BMPCs, which was reversed by Ang-(1-7). In diabetic BMPCs, SDF and angiopoietin-1 were upregulated by Ang-(1-7), and in cavernosum, VEGFR1, Tie-2, and SDF were upregulated and angiopoietin-2 was down-regulated. CONCLUSIONS: Ang-(1-7) stimulates angiogenic function of cavernosum in diabetes via its stimulating effects on both cavernosal microvasculature and BMPCs.


Subject(s)
Angiotensin I/pharmacology , Bone Marrow Cells/drug effects , Diabetes Mellitus, Experimental/physiopathology , Microvessels/drug effects , Neovascularization, Pathologic/prevention & control , Penis/drug effects , Peptide Fragments/pharmacology , Animals , Diabetes Mellitus, Type 1/physiopathology , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Penis/blood supply , Penis/metabolism , Proto-Oncogene Mas , Real-Time Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/pharmacology
12.
PLoS One ; 9(4): e93965, 2014.
Article in English | MEDLINE | ID: mdl-24713821

ABSTRACT

We hypothesized that endothelial progenitor cells derived from individuals with diabetes would exhibit functional defects including inability to respond to hypoxia and altered paracrine/autocrine function that would impair the angiogenic potential of these cells. Circulating mononuclear cells isolated from diabetic (n = 69) and nondiabetic (n = 46) individuals were used to grow endothelial colony forming cells (ECFC), early endothelial progenitor cells (eEPCs) and isolate CD34+ cells. ECFCs and eEPCs were established from only 15% of the diabetic individuals tested thus directing our main effort toward examination of CD34+ cells. CD34+ cells were plated in basal medium to obtain cell-free conditioned medium (CM). In CM derived from CD34+ cells of diabetic individuals (diabetic-CM), the levels of stem cell factor, hepatocyte growth factor, and thrombopoietin were lower, and IL-1ß and tumor necrosis factor (TNFα) levels were higher than CM derived from nondiabetic individuals (nondiabetic-CM). Hypoxia did not upregulate HIF1α in CD34+ cells of diabetic origin. Migration and proliferation of nondiabetic CD34+ cells toward diabetic-CM were lower compared to nondiabetic-CM. Attenuation of pressure-induced constriction, potentiation of bradykinin relaxation, and generation of cGMP and cAMP in arterioles were observed with nondiabetic-CM, but not with diabetic-CM. Diabetic-CM failed to induce endothelial tube formation from vascular tissue. These results suggest that diabetic subjects with microvascular complications exhibit severely limited capacity to generate ex-vivo expanded endothelial progenitor populations and that the vasoreparative dysfunction observed in diabetic CD34+ cells is due to impaired autocrine/paracrine function and reduced sensitivity to hypoxia.


Subject(s)
Antigens, CD34/metabolism , Diabetes Mellitus, Type 2/metabolism , Endothelial Cells/metabolism , Hypoxia/metabolism , Neovascularization, Physiologic/physiology , Stem Cells/metabolism , Adult , Aged , Cells, Cultured , Hepatocyte Growth Factor/metabolism , Humans , Male , Middle Aged , Stem Cell Factor/metabolism , Thrombopoietin/metabolism , Tumor Necrosis Factor-alpha/metabolism
13.
Eur J Pharmacol ; 705(1-3): 42-8, 2013 Apr 05.
Article in English | MEDLINE | ID: mdl-23454521

ABSTRACT

The use of opioids, which achieve therapeutic analgesia through activation of µ-opioid receptors, are limited in the management of chronic pain by adverse effects including tolerance and addiction. Optogenetics is an emerging approach of designing molecular targets that can produce cell-specific receptor-mediated analgesia with minimal side effects. Here we report the design and functional characterization of a chimeric µ-opioid receptor that could be photoactivated to trigger intracellular signaling. A prototype optoactive µ-opioid receptor (optoMOR) was designed by replacing the intracellular domains from rhodopsin with those of the native µ-opioid receptor and was transiently expressed in human embryonic kidney (HEK293) cells. Expression and distribution of the protein were confirmed by immunocytochemistry. The signal-transduction mechanisms induced by photoactivation of the optoMOR were evaluated and compared with the native µ-opioid receptor stimulation by an agonist, D-Ala(2), N-MePhe(4), Gly-ol-enkephalin (DAMGO). Cells were depolarized by extracellular potassium and the depolarization-induced calcium (Ca(2+)) influx was quantified by using Fura-2 imaging. The forskolin-stimulated adenylate cyclase/cAMP cascade was evaluated by ELISA or western blotting of brain-derived neurotrophic factor (BDNF) and the phosphorylation of cAMP response element binding protein (CREB). The optoMOR protein distribution was observed intracellularly and on the plasma membrane similar to the native µ-opioid receptor in HEK293 cells. Photoactivation of optoMOR decreased the Ca(2+) influx and inhibited the forskolin-induced cAMP generation, activation of CREB, and BDNF levels in optoMOR-expressing cells similar to the activation of native µ-opioid receptor by DAMGO. Thus the current study has accomplished the design of a prototype optoMOR and characterized the cellular signaling mechanisms activated by light stimulation of this receptor.


Subject(s)
Receptors, Opioid, mu/genetics , Analgesics, Opioid/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , HEK293 Cells , Humans , Optogenetics , Photic Stimulation , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Rhodopsin/genetics
14.
Diabetes ; 62(4): 1258-69, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23230080

ABSTRACT

We tested the hypothesis that activation of the protective arm of the renin angiotensin system, the angiotensin-converting enzyme 2 (ACE2)/angiotensin-(1-7) [Ang-(1-7)]/Mas receptor axis, corrects the vasoreparative dysfunction typically seen in the CD34(+) cells isolated from diabetic individuals. Peripheral blood CD34(+) cells from patients with diabetes were compared with those of nondiabetic controls. Ang-(1-7) restored impaired migration and nitric oxide bioavailability/cGMP in response to stromal cell-derived factor and resulted in a decrease in NADPH oxidase activity. The survival and proliferation of CD34(+) cells from diabetic individuals were enhanced by Ang-(1-7) in a Mas/phosphatidylinositol 3-kinase (PI3K)/Akt-dependent manner. ACE2 expression was lower, and ACE2 activators xanthenone and diminazine aceturate were less effective in inducing the migration in cells from patients with diabetes compared with controls. Ang-(1-7) overexpression by lentiviral gene modification restored both the in vitro vasoreparative functions of diabetic cells and the in vivo homing efficiency to areas of ischemia. A cohort of patients who remained free of microvascular complications despite having a history of longstanding inadequate glycemic control had higher expression of ACE2/Mas mRNA than patients with diabetes with microvascular complications matched for age, sex, and glycemic control. Thus, ACE2/Ang-(1-7)\Mas pathway activation corrects existing diabetes-induced CD34(+) cell dysfunction and also confers protection from development of this dysfunction.


Subject(s)
Angiotensin I/metabolism , Diabetes Mellitus/metabolism , Endothelial Cells/physiology , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Stem Cells/physiology , Adult , Angiotensin I/genetics , Angiotensin-Converting Enzyme 2 , Animals , Antigens, CD34/genetics , Antigens, CD34/metabolism , Case-Control Studies , Cohort Studies , Female , Gene Expression Regulation , Humans , Male , Mice , Middle Aged , Peptide Fragments/genetics , Peptidyl-Dipeptidase A/genetics , Proto-Oncogene Mas , Renin-Angiotensin System/physiology
15.
PLoS One ; 7(7): e39398, 2012.
Article in English | MEDLINE | ID: mdl-22792172

ABSTRACT

Previously, we showed that insulin growth factor (IGF)-1 binding protein-3 (IGFBP-3), independent of IGF-1, reduces pathological angiogenesis in a mouse model of the oxygen-induced retinopathy (OIR). The current study evaluates novel endothelium-dependent functions of IGFBP-3 including blood retinal barrier (BRB) integrity and vasorelaxation. To evaluate vascular barrier function, either plasmid expressing IGFBP-3 under the regulation of an endothelial-specific promoter or a control plasmid was injected into the vitreous humor of mouse pups (P1) and compared to the non-injected eyes of the same pups undergoing standard OIR protocol. Prior to sacrifice, the mice were given an injection of horseradish peroxidase (HRP). IGFBP-3 plasmid-injected eyes displayed near-normal vessel morphology and enhanced vascular barrier function. Further, in vitro IGFBP-3 protects retinal endothelial cells from VEGF-induced loss of junctional integrity by antagonizing the dissociation of the junctional complexes. To assess the vasodilatory effects of IGFBP-3, rat posterior cerebral arteries were examined in vitro. Intraluminal IGFBP-3 decreased both pressure- and serotonin-induced constrictions by stimulating nitric oxide (NO) release that were blocked by L-NAME or scavenger receptor-B1 neutralizing antibody (SRB1-Ab). Both wild-type and IGF-1-nonbinding mutant IGFBP-3 (IGFBP-3NB) stimulated eNOS activity/NO release to a similar extent in human microvascular endothelial cells (HMVECs). NO release was neither associated with an increase in intracellular calcium nor decreased by Ca(2+)/calmodulin-dependent protein kinase II (CamKII) blockade; however, dephosphorylation of eNOS-Thr(495) was observed. Phosphatidylinositol 3-kinase (PI3K) activity and Akt-Ser(473) phosphorylation were both increased by IGFBP-3 and selectively blocked by the SRB1-Ab or PI3K blocker LY294002. In conclusion, IGFBP-3 mediates protective effects on BRB integrity and mediates robust NO release to stimulate vasorelaxation via activation of SRB1. This response is IGF-1- and calcium-independent, but requires PI3K/Akt activation, suggesting that IGFBP-3 has novel protective effects on retinal and systemic vasculature and may be a therapeutic candidate for ocular complications such as diabetic retinopathy.


Subject(s)
Blood-Retinal Barrier/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Animals , Blood-Retinal Barrier/pathology , Cadherins/genetics , Cadherins/metabolism , Calcium/metabolism , Cerebral Arteries/drug effects , Cerebral Arteries/metabolism , Claudin-5/genetics , Claudin-5/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fura-2 , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor I/metabolism , Intercellular Junctions/drug effects , Intercellular Junctions/genetics , Intercellular Junctions/metabolism , Male , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Nitric Oxide , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Retinal Diseases/genetics , Retinal Diseases/metabolism , Retinal Vessels/drug effects , Retinal Vessels/metabolism , Scavenger Receptors, Class B/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/pharmacology
16.
Invest Ophthalmol Vis Sci ; 52(8): 5093-104, 2011 Jul 07.
Article in English | MEDLINE | ID: mdl-21676908

ABSTRACT

PURPOSE: The vasodegenerative phase of diabetic retinopathy is likely caused by endothelial dysfunction and reduced endothelial repair. Migration of endothelial progenitor cells (EPCs) into areas of vascular injury is critical to vascular repair. This key function, often defective in diabetes, is largely mediated by nitric oxide (NO), which is known to be inactivated by superoxide produced by NADPH oxidase. The authors tested the hypothesis that either increasing eNOS expression or inhibiting NADPH oxidase would restore the reparative function in diabetic EPCs. METHODS: Peripheral blood was obtained from healthy (n = 27) and diabetic (n = 31) persons, and CD34(+) cells were isolated. Expression and activation of eNOS and NADPH oxidase and intracellular levels of NO, superoxide, and peroxynitrite were evaluated. cGMP production and migration to SDF-1α were also determined. Reparative function was evaluated in a mouse model of retinal ischemia-reperfusion injury. RESULTS: Diabetic EPCs demonstrate reduced eNOS expression and decreased NO bioavailability and migration in response to SDF-1α. Increasing eNOS expression in diabetic cells by AVE3085 resulted in increased peroxynitrite levels and, therefore, did not enhance NO-mediated functions in vitro and in vivo. Expression of Nox2, NADPH oxidase activity, and superoxide levels were higher in diabetic than in nondiabetic EPCs. Pretreatment with apocynin or gp91ds-tat increased NO bioavailability without increasing eNOS activity in response to SDF-1α. Ex vivo NADPH oxidase inhibition in diabetic cells restored migratory function in vitro and enhanced their homing to ischemic retinal vasculature in vivo. CONCLUSIONS: The NADPH oxidase system is a promising target for correcting vasoreparative dysfunction in diabetic EPCs.


Subject(s)
Antigens, CD34/metabolism , Diabetic Retinopathy/blood , Endothelium, Vascular/enzymology , Enzyme Inhibitors/pharmacology , NADPH Oxidases/antagonists & inhibitors , Reperfusion Injury/physiopathology , Acetophenones/pharmacology , Adult , Animals , Benzodioxoles/pharmacology , Chemokine CXCL12/metabolism , Cyclic GMP/metabolism , Disease Models, Animal , Female , Glycoproteins/pharmacology , Humans , Indans/pharmacology , Male , Mice , Middle Aged , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Peroxynitrous Acid/metabolism , Reperfusion Injury/enzymology , Reverse Transcriptase Polymerase Chain Reaction , Superoxides/metabolism , Young Adult
17.
Endocrinology ; 152(7): 2786-96, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21558316

ABSTRACT

The peptide hormone relaxin is a potent vasodilator with therapeutic potential in diseases complicated by vasoconstriction, including heart failure. However, the molecular mediators and magnitude of vasodilation may vary according to duration of exposure and artery type. The objective of these studies was to determine mechanisms of rapid (within minutes) relaxin-induced vasodilation and to examine whether relaxin dilates arteries from different animal species and vascular beds. Rat and mouse small renal, rat mesenteric, and human sc arteries were isolated, mounted in a pressure arteriograph, and treated with recombinant human relaxin (rhRLX; 1-100 ng/ml) after preconstriction with phenylephrine. Rat and mouse small renal as well as human sc arteries dilated in response to rhRLX, whereas rat mesenteric arteries did not. Endothelial removal or pretreatment with l-N(G)-monomethyl arginine (L-NMMA) abolished rapid relaxin-induced vasodilation; phosphatidylinositol-3-kinase (PI3K) inhibitors also prevented it. In cultured human endothelial cells, rhRLX stimulated nitric oxide (assessed using 4-amino-5-methylamino-2'7'-difluorofluorescein) as well as Akt and endothelial NO synthase (eNOS) phosphorylation by Western blotting but not increases in intracellular calcium (evaluated by fura-2). NO production was attenuated by inhibition of Gα(i/o) and Akt (using pertussis toxin and the allosteric inhibitor MK-2206, respectively), PI3K, and NOS. Finally, the dilatory effect of rhRLX in rat small renal arteries was unexpectedly potentiated, rather than inhibited, by pretreatment with the vascular endothelial growth factor receptor inhibitor SU5416. We conclude that relaxin rapidly dilates select arteries across a range of species. The mechanism appears to involve endothelial Gα(i/o) protein coupling to PI3K, Akt, and eNOS but not vascular endothelial growth factor receptor transactivation or increased calcium.


Subject(s)
Kidney/blood supply , Nitric Oxide/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Relaxin/physiology , Subcutaneous Tissue/blood supply , Vasodilation , Adult , Angiogenesis Inhibitors/pharmacology , Animals , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Enzyme Inhibitors/pharmacology , Female , Humans , In Vitro Techniques , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Organ Specificity , Phosphoinositide-3 Kinase Inhibitors , Rats , Rats, Long-Evans , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Species Specificity , Vasodilation/drug effects
18.
Diabetes ; 59(8): 2010-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20460428

ABSTRACT

OBJECTIVE: Peripheral blood CD34(+) cells from diabetic patients demonstrate reduced vascular reparative function due to decreased proliferation and diminished migratory prowess, largely resulting from decreased nitric oxide (NO) bioavailability. The level of TGF-beta, a key factor that modulates stem cell quiescence, is increased in the serum of type 2 diabetic patients. We asked whether transient TGF-beta1 inhibition in CD34(+) cells would improve their reparative ability. RESEARCH DESIGN AND METHODS: To inhibit TGF-beta1 protein expression, CD34(+) cells were treated ex vivo with antisense phosphorodiamidate morpholino oligomers (TGF-beta1-PMOs) and analyzed for cell surface CXCR4 expression, cell survival in the absence of added growth factors, SDF-1-induced migration, NO release, and in vivo retinal vascular reparative ability. RESULTS: TGF-beta1-PMO treatment of diabetic CD34(+) cells resulted in increased expression of CXCR4, enhanced survival in the absence of growth factors, and increased migration and NO release as compared with cells treated with control PMO. Using a retinal ischemia reperfusion injury model in mice, we observed that recruitment of diabetic CD34(+) cells to injured acellular retinal capillaries was greater after TGF-beta1-PMO treatment compared with control PMO-treated cells. CONCLUSIONS: Transient inhibition of TGF-beta1 may represent a promising therapeutic strategy for restoring the reparative capacity of dysfunctional diabetic CD34(+) cells.


Subject(s)
Diabetes Mellitus/physiopathology , Diabetic Angiopathies/prevention & control , Hematopoietic Stem Cells/physiology , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta/metabolism , Animals , Antigens, CD34/metabolism , Antigens, CD34/physiology , Capillaries/physiopathology , Cell Survival , Diabetic Retinopathy/physiopathology , Flow Cytometry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Mice , Morpholines/pharmacology , Morpholines/therapeutic use , Morpholinos , Nitric Oxide/metabolism , Receptors, CXCR4/genetics , Reperfusion Injury/physiopathology , Transforming Growth Factor beta1/metabolism
19.
Invest Ophthalmol Vis Sci ; 51(10): 5240-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20435587

ABSTRACT

PURPOSE: Dysfunction of endothelial nitric oxide synthase (eNOS) has been implicated in the pathogenesis of diabetic vascular complications. This study was undertaken to determine the role of eNOS in the development of diabetic retinopathy (DR), by investigating the functional consequences of its deficiency in the diabetic state. METHODS: Diabetes was induced in eNOS-knockout (eNOS(-/-)) and C57B/6 mice by streptozotocin (STZ) injection. Retinal vasculature was evaluated by albumin extravasation, to quantitatively measure vascular permeability, and by trypsin-digested retinal vascular preparations, to quantify acellular capillaries. Gliosis was evaluated by immunofluorescent techniques. Retinal capillary basement membrane thickness was assessed by transmission electron microscopy. Total retinal nitric oxide level was assessed by measuring nitrate/nitrite using a fluorometric-based assay, iNOS expression was examined by real-time PCR. RESULTS: Diabetic eNOS(-/-) mice exhibit more severe retinal vascular permeability than age-matched diabetic C57BL/6 mice, detectable as early as 3 weeks after diabetes induction. Diabetic eNOS(-/-) mice also show earlier onset and an increased number of acellular capillaries, sustained gliosis, and increased capillary basement membrane thickness. Total nitric oxide (NO) level was also increased, concomitant with elevated iNOS expression in diabetic eNOS(-/-) retina. CONCLUSIONS: Diabetic eNOS(-/-) mice exhibit A significantly wider range of advanced retinal vascular complications than the age-matched diabetic C57BL/6 mice, supporting the notion that eNOS-derived NO plays an essential role in retinal vascular function. This mouse model also faithfully replicates many of the hallmarks of vascular changes associated with human retinopathy, thus providing a unique model to aid in understanding the pathologic mechanisms of and to develop effective therapeutic strategies for diabetic retinopathy.


Subject(s)
Diabetes Mellitus, Experimental/enzymology , Diabetic Retinopathy/enzymology , Nitric Oxide Synthase Type III/physiology , Retina/enzymology , Albumins/metabolism , Animals , Capillary Permeability , Diabetes Mellitus, Experimental/physiopathology , Diabetic Retinopathy/physiopathology , Female , Fluorescein Angiography , Fluorescent Antibody Technique, Indirect , Glial Fibrillary Acidic Protein/metabolism , Gliosis/diagnosis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/physiology , Nitric Oxide Synthase Type II/genetics , Retina/physiopathology , Retinal Vessels/pathology , Reverse Transcriptase Polymerase Chain Reaction
20.
Circ Res ; 106(5): 854-69, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20299675

ABSTRACT

The discovery of endothelial progenitor cells (EPCs) in human peripheral blood advanced the field of cell-based therapeutics for many pathological conditions. Despite the lack of agreement about the existence and characteristics of EPCs, autologous EPC populations represent a novel treatment option for complications requiring therapeutic revascularization and vascular repair. Patients with diabetic complications represent a population of patients that may benefit from cellular therapy yet their broadly dysfunctional cells may limit the feasibility of this approach. Diabetic EPCs have decreased migratory prowess and reduced proliferative capacity and an altered cytokine/growth factor secretory profile that can accelerate deleterious repair mechanisms rather than support proper vascular repair. Furthermore, the diabetic environment poses additional challenges for the autologous transplantation of cells. The present review is focused on correcting diabetic EPC dysfunction and the challenges involved in the application of cell-based therapies for treatment of diabetic vascular complications. In addition, ex vivo and in vivo functional manipulation(s) of EPCs to overcome these hurdles are discussed.


Subject(s)
Diabetes Complications/therapy , Endothelial Cells/transplantation , Endothelium, Vascular/physiopathology , Genetic Therapy , Stem Cell Transplantation , Animals , Cell Movement , Cell Proliferation , Diabetes Complications/genetics , Diabetes Complications/physiopathology , Genetic Therapy/adverse effects , Humans , Neovascularization, Physiologic , Paracrine Communication , Stem Cell Transplantation/adverse effects , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...