Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 232
Filter
1.
Am J Physiol Renal Physiol ; 327(1): F103-F112, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38779750

ABSTRACT

α-1-Microglobulin (A1M) is a circulating glycoprotein with antioxidant, heme-binding, and mitochondrial protection properties. The investigational drug RMC-035, a modified therapeutic A1M protein, was assessed for biodistribution and pharmacological activity in a broad set of in vitro and in vivo experiments, supporting its clinical development. Efficacy and treatment posology were assessed in various models of kidney ischemia and reperfusion injury (IRI). Real-time glomerular filtration rate (GFR), functional renal biomarkers, tubular injury biomarkers (NGAL and KIM-1), and histopathology were evaluated. Fluorescently labeled RMC-035 was used to assess biodistribution. RMC-035 demonstrated consistent and reproducible kidney protection in rat IRI models as well as in a model of IRI imposed on renal impairment and in a mouse IRI model, where it reduced mortality. Its pharmacological activity was most pronounced with combined dosing pre- and post-ischemia and weaker with either pre- or post-ischemia dosing alone. RMC-035 rapidly distributed to the kidneys via glomerular filtration and selective luminal uptake by proximal tubular cells. IRI-induced expression of kidney heme oxygenase-1 was inhibited by RMC-035, consistent with its antioxidative properties. RMC-035 also dampened IRI-associated inflammation and improved mitochondrial function, as shown by tubular autofluorescence. Taken together, the efficacy of RMC-035 is congruent with its targeted mechanism(s) and biodistribution profile, supporting its further clinical evaluation as a novel kidney-protective therapy.NEW & NOTEWORTHY A therapeutic A1M protein variant (RMC-035) is currently in phase 2 clinical development for renal protection in patients undergoing open-chest cardiac surgery. It targets several key pathways underlying kidney injury in this patient group, including oxidative stress, heme toxicity, and mitochondrial dysfunction. RMC-035 is rapidly eliminated from plasma, distributing to kidney proximal tubules, and demonstrates dose-dependent efficacy in numerous models of ischemia-reperfusion injury, particularly when administered before ischemia. These results support its continued clinical evaluation.


Subject(s)
Alpha-Globulins , Kidney , Reperfusion Injury , Animals , Reperfusion Injury/pathology , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Reperfusion Injury/drug therapy , Alpha-Globulins/metabolism , Alpha-Globulins/pharmacology , Male , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Disease Models, Animal , Glomerular Filtration Rate/drug effects , Mice, Inbred C57BL , Humans , Mice , Heme Oxygenase-1/metabolism , Rats , Rats, Sprague-Dawley , Acute Kidney Injury/pathology , Acute Kidney Injury/metabolism , Acute Kidney Injury/drug therapy , Acute Kidney Injury/prevention & control , Tissue Distribution
2.
Neurotherapeutics ; 21(3): e00341, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38453562

ABSTRACT

Therapeutic hypothermia is the standard of care for hypoxic-ischemic (HI) encephalopathy. Inter-alpha Inhibitor Proteins (IAIPs) attenuate brain injury after HI in neonatal rats. Human (h) IAIPs (60 â€‹mg/kg) or placebo (PL) were given 15 â€‹min, 24 and 48 â€‹h to postnatal (P) day-7 rats after carotid ligation and 8% oxygen for 90 â€‹min with (30 â€‹°C) and without (36 â€‹°C) exposure to hypothermia 1.5 â€‹h after HI for 3 â€‹h. Hemispheric volume atrophy (P14) and neurobehavioral tests including righting reflex (P8-P10), small open field (P13-P14), and negative geotaxis (P14) were determined. Hemispheric volume atrophy in males was reduced (P â€‹< â€‹0.05) by 41.9% in the normothermic-IAIP and 28.1% in the hypothermic-IAIP compared with the normothermic-PL group, and in females reduced (P â€‹< â€‹0.05) by 30.3% in the normothermic-IAIP, 45.7% in hypothermic-PL, and 55.2% in hypothermic-IAIP compared with the normothermic-PL group after HI. Hypothermia improved (P â€‹< â€‹0.05) the neuroprotective effects of hIAIPs in females. The neuroprotective efficacy of hIAIPs was comparable to hypothermia in female rats (P â€‹= â€‹0.183). Treatment with hIAIPs, hypothermia, and hIAIPs with hypothermia decreased (P â€‹< â€‹0.05) the latency to enter the peripheral zone in the small open field test in males. We conclude that hIAIPs provide neuroprotection from HI brain injury that is comparable to the protection by hypothermia, hypothermia increases the effects of hIAIPs in females, and hIAIPs and hypothermia exhibit some sex-related differential effects.


Subject(s)
Alpha-Globulins , Hypothermia, Induced , Hypoxia-Ischemia, Brain , Neuroprotective Agents , Animals , Female , Humans , Male , Rats , Alpha-Globulins/metabolism , Alpha-Globulins/pharmacology , Animals, Newborn , Hypothermia, Induced/methods , Hypoxia-Ischemia, Brain/therapy , Hypoxia-Ischemia, Brain/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats, Sprague-Dawley
3.
Int J Mol Sci ; 21(16)2020 Aug 13.
Article in English | MEDLINE | ID: mdl-32823731

ABSTRACT

Oxidative stress is associated with many renal disorders, both acute and chronic, and has also been described to contribute to the disease progression. Therefore, oxidative stress is a potential therapeutic target. The human antioxidant α1-microglobulin (A1M) is a plasma and tissue protein with heme-binding, radical-scavenging and reductase activities. A1M can be internalized by cells, localized to the mitochondria and protect mitochondrial function. Due to its small size, A1M is filtered from the blood into the glomeruli, and taken up by the renal tubular epithelial cells. A1M has previously been described to reduce renal damage in animal models of preeclampsia, radiotherapy and rhabdomyolysis, and is proposed as a pharmacological agent for the treatment of kidney damage. In this paper, we examined the in vitro protective effects of recombinant human A1M (rA1M) in human proximal tubule epithelial cells. Moreover, rA1M was found to protect against heme-induced cell-death both in primary cells (RPTEC) and in a cell-line (HK-2). Expression of stress-related genes was upregulated in both cell cultures in response to heme exposure, as measured by qPCR and confirmed with in situ hybridization in HK-2 cells, whereas co-treatment with rA1M counteracted the upregulation. Mitochondrial respiration, analyzed with the Seahorse extracellular flux analyzer, was compromised following exposure to heme, but preserved by co-treatment with rA1M. Finally, heme addition to RPTE cells induced an upregulation of the endogenous cellular expression of A1M, via activation of the nuclear factor erythroid 2-related factor 2 (Nrf2)-pathway. Overall, data suggest that A1M/rA1M protects against stress-induced damage to tubule epithelial cells that, at least partly, can be attributed to maintaining mitochondrial function.


Subject(s)
Alpha-Globulins/pharmacology , Epithelial Cells/pathology , Heme/toxicity , Kidney Tubules, Proximal/pathology , Protective Agents/pharmacology , Apoptosis/drug effects , Cell Line , Cytoprotection/drug effects , HSP70 Heat-Shock Proteins/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Stress, Physiological/drug effects , Up-Regulation/drug effects
4.
J Cereb Blood Flow Metab ; 40(5): 1090-1102, 2020 05.
Article in English | MEDLINE | ID: mdl-31234704

ABSTRACT

Circulating levels of inter-alpha inhibitor proteins change dramatically in acute inflammatory disorders, which suggest an important contribution to the immunomodulatory system. Human blood-derived inter-alpha inhibitor proteins are neuroprotective and improve survival of neonatal mice exposed to lipopolysaccharide. Lipopolysaccharide augments inflammatory conditions and disrupts the blood-brain barrier. There is a paucity of therapeutic strategies to treat blood-brain barrier dysfunction, and the neuroprotective effects of human blood-derived inter-alpha inhibitor proteins are not fully understood. To examine the therapeutic potential of inter-alpha inhibitor proteins, we administered human blood-derived inter-alpha inhibitor proteins to male and female CD-1 mice after lipopolysaccharide exposure and quantified blood-brain barrier permeability of intravenously injected 14C-sucrose and 99mTc-albumin. We hypothesized that human blood-derived inter-alpha inhibitor protein treatment would attenuate lipopolysaccharide-induced blood-brain barrier disruption and associated inflammation. Lipopolysaccharide increased blood-brain barrier permeability to both 14C-sucrose and 99mTc-albumin, but human blood-derived inter-alpha inhibitor protein treatment only attenuated increases in 14C-sucrose blood-brain barrier permeability in male mice. Lipopolysaccharide stimulated a more robust elevation of male serum inter-alpha inhibitor protein concentration compared to the elevation measured in female serum. Lipopolysaccharide administration also increased multiple inflammatory factors in serum and brain tissue, including interleukin 6. Human blood-derived inter-alpha inhibitor protein treatment downregulated serum interleukin 6 levels, which were inversely correlated with serum inter-alpha inhibitor protein concentration. We conclude that inter-alpha inhibitor proteins may be neuroprotective through mechanisms of blood-brain barrier disruption associated with systemic inflammation.


Subject(s)
Alpha-Globulins/pharmacology , Blood-Brain Barrier/drug effects , Capillary Permeability/drug effects , Interleukin-6/metabolism , Animals , Down-Regulation , Female , Humans , Inflammation/chemically induced , Inflammation/metabolism , Lipopolysaccharides/toxicity , Male , Mice , Neuroprotective Agents/pharmacology
5.
Sci Rep ; 9(1): 8561, 2019 06 12.
Article in English | MEDLINE | ID: mdl-31189914

ABSTRACT

Preeclampsia is a human placental disorder affecting 2-8% of pregnancies worldwide annually, with hypertension and proteinuria appearing after 20 weeks of gestation. The underlying cause is believed to be incomplete trophoblast invasion of the maternal spiral arteries during placentation in the first trimester, resulting in oxidative and nitrative stress as well as maternal inflammation and organ alterations. In the Storkhead box 1 (STOX1) preeclampsia mouse model, pregnant females develop severe and early onset manifestations as seen in human preeclampsia e.g. gestational hypertension, proteinuria, and organ alterations. Here we aimed to evaluate the therapeutic potential of human recombinant alpha-1 microglobulin (rA1M) to alleviate the manifestations observed. Human rA1M significantly reduced the hypertension during gestation and significantly reduced the level of hypoxia and nitrative stress in the placenta. In addition, rA1M treatment reduced cellular damage in both placenta and kidneys, thereby protecting the tissue and improving their function. This study confirms that rA1M has the potential as a therapeutic drug in preeclampsia, and likely also in other pathological conditions associated with oxidative stress, by preserving normal organ function.


Subject(s)
Alpha-Globulins/pharmacology , Oxidative Stress/drug effects , Pre-Eclampsia , Animals , Disease Models, Animal , Female , Humans , Mice , Mice, Transgenic , Oxidative Stress/genetics , Pre-Eclampsia/drug therapy , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Pregnancy , Recombinant Proteins/pharmacology
6.
J Neuroinflammation ; 16(1): 122, 2019 Jun 07.
Article in English | MEDLINE | ID: mdl-31174551

ABSTRACT

BACKGROUND: Germinal matrix intraventricular hemorrhage (GM-IVH) is associated with cerebro-cerebellar damage in very preterm infants, leading to neurodevelopmental impairment. Penetration, from the intraventricular space, of extravasated red blood cells and extracellular hemoglobin (Hb), to the periventricular parenchyma and the cerebellum has been shown to be causal in the development of brain injury following GM-IVH. Furthermore, the damage has been described to be associated with the cytotoxic nature of extracellular Hb-metabolites. To date, there is no therapy available to prevent infants from developing either hydrocephalus or serious neurological disability. Mechanisms previously described to cause brain damage following GM-IVH, i.e., oxidative stress and Hb-metabolite toxicity, suggest that the free radical and heme scavenger α1-microglobulin (A1M) may constitute a potential neuroprotective intervention. METHODS: Using a preterm rabbit pup model of IVH, where IVH was induced shortly after birth in pups delivered by cesarean section at E29 (3 days prior to term), we investigated the brain distribution of recombinant A1M (rA1M) following intracerebroventricular (i.c.v.) administration at 24 h post-IVH induction. Further, short-term functional protection of i.c.v.-administered human A1M (hA1M) following IVH in the preterm rabbit pup model was evaluated. RESULTS: Following i.c.v. administration, rA1M was distributed in periventricular white matter regions, throughout the fore- and midbrain and extending to the cerebellum. The regional distribution of rA1M was accompanied by a high co-existence of positive staining for extracellular Hb. Administration of i.c.v.-injected hA1M was associated with decreased structural tissue and mitochondrial damage and with reduced mRNA expression for proinflammatory and inflammatory signaling-related genes induced by IVH in periventricular brain tissue. CONCLUSIONS: The results of this study indicate that rA1M/hA1M is a potential candidate for neuroprotective treatment following preterm IVH.


Subject(s)
Alpha-Globulins/pharmacology , Brain/drug effects , Brain/pathology , Cerebral Intraventricular Hemorrhage/etiology , Cerebral Intraventricular Hemorrhage/pathology , Free Radical Scavengers/pharmacology , Premature Birth , Animals , Animals, Newborn , Female , Humans , Male , Pregnancy , Rabbits , Random Allocation
7.
Lung ; 197(3): 361-369, 2019 06.
Article in English | MEDLINE | ID: mdl-31028466

ABSTRACT

PURPOSE: Vascular endothelial cells demonstrate severe injury in sepsis, and a reduction in endothelial inflammation would be beneficial. Inter-α-Inhibitor (IαI) is a family of abundant plasma proteins with anti-inflammatory properties and has been investigated in human and animal sepsis with encouraging results. We hypothesized that IαI may protect endothelia from sepsis-related inflammation. METHODS: IαI-deficient or sufficient mice were treated with endotoxin or underwent complement-induced lung injury. VCAM-1 and ICAM-1 expression was measured in blood and lung as marker of endothelial activation. Human endothelia were exposed to activated complement C5a with or without IαI. Blood from human sepsis patients was examined for VCAM-1 and ICAM-1 and levels were correlated with blood levels of IαI. RESULTS: IαI-deficient mice showed increased endothelial activation in endotoxin/sepsis- and complement-induced lung injury models. In vitro, levels of endothelial pro-inflammatory cytokines and cell growth factors induced by activated complement C5a were significantly decreased in the presence of IαI. This effect was associated with decreased ERK and NFκB activation. IαI levels were inversely associated with VCAM-1 and ICAM-1 levels in a human sepsis cohort. CONCLUSIONS: IαI ameliorates endothelial inflammation and may be beneficial as a treatment of sepsis.


Subject(s)
Acute Lung Injury/immunology , Alpha-Globulins/immunology , Endothelial Cells/immunology , Endothelium, Vascular/immunology , Lung/immunology , Sepsis/immunology , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Alpha-Globulins/deficiency , Alpha-Globulins/metabolism , Alpha-Globulins/pharmacology , Animals , Complement C5a/immunology , Complement C5a/pharmacology , Disease Models, Animal , E-Selectin/immunology , E-Selectin/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endotoxins/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Inflammation , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/metabolism , Lung/drug effects , Lung/metabolism , Lung/pathology , MAP Kinase Signaling System , Mice , NF-kappa B/drug effects , NF-kappa B/immunology , NF-kappa B/metabolism , Sepsis/genetics , Sepsis/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology , Vascular Cell Adhesion Molecule-1/metabolism
8.
J Nucl Med ; 60(11): 1600-1604, 2019 11.
Article in English | MEDLINE | ID: mdl-30926650

ABSTRACT

Treatment of neuroendocrine tumors with 177Lu-octreotate results in prolonged survival and improved quality of life for the patient. However, the treatment is today limited by side effects on kidney and bone marrow, and complete tumor remission is rarely seen. A possible way to minimize dose-limiting toxicity and to optimize this treatment method is to use radioprotectors in conjunction with radiotherapy. A recombinant form of α1-microglobulin (rA1M) was recently shown to preserve kidney structure and function after 177Lu-octreotate injection in mice and was suggested as a radioprotector in peptide receptor radionuclide therapy. The aims of this work were to investigate the influence of rA1M on the in vivo biokinetics of 177Lu-octreotate, with a focus on tumor tissue, and to study the impact of rA1M on the therapeutic response in tumor tissue subjected to 177Lu-octreotate treatment. Methods: The biodistribution of 177Lu-octreotate was examined in BALB/c nude mice with GOT2 tumors 1-168 h after injection with either 177Lu-octreotate or coadministration of 177Lu-octreotate and rA1M. The effects of rA1M on the tumor response after 177Lu-octreotate treatment were studied in BALB/c nude mice with GOT1 tumors. Three groups of mice were administered rA1M, 177Lu-octreotate, or both. Another group served as untreated controls. Tumor volume was measured to follow the treatment effects. Results: No statistically significant difference in biodistribution of 177Lu was observed between the groups receiving 177Lu-octreotate or coinjection of 177Lu-octreotate and rA1M. The therapy study showed a decrease in mean tumor volume during the first 2 wk for both the 177Lu-octreotate group and the coadministration group, followed by tumor regrowth. No statistically significant difference between the groups was found. Conclusion: rA1M did not negatively impact absorbed dose to tumor or therapeutic response in combination with 177Lu-octreotate and may be a promising kidney protector during 177Lu-octreotate treatment of patients with neuroendocrine tumors.


Subject(s)
Alpha-Globulins/pharmacology , Kidney/drug effects , Kidney/radiation effects , Neuroendocrine Tumors/radiotherapy , Octreotide/analogs & derivatives , Recombinant Proteins/pharmacology , Animals , Biological Transport/drug effects , Cytoprotection/drug effects , Female , Kidney/metabolism , Kinetics , Mice , Mice, Inbred BALB C , Neuroendocrine Tumors/metabolism , Octreotide/adverse effects , Octreotide/metabolism , Octreotide/pharmacokinetics , Octreotide/therapeutic use , Organs at Risk/radiation effects , Tissue Distribution/drug effects
9.
Exp Neurol ; 317: 244-259, 2019 07.
Article in English | MEDLINE | ID: mdl-30914159

ABSTRACT

Hypoxic-ischemic (HI) brain injury is one of the most common neurological problems occurring in the perinatal period. Hypothermia is the only approved intervention for neonatal HI encephalopathy. However, this treatment is only partially protective, has a narrow therapeutic time window after birth and only can be used to treat full-term infants. Consequently, additional therapies are critically needed. Inflammation is an important contributing factor to the evolution of HI brain injury in neonates. Inter-alpha Inhibitor Proteins (IAIPs) are immunomodulatory proteins with anti-inflammatory properties. We have previously shown that IAIPs reduce neuronal cell death and improve behavioral outcomes when given after carotid artery ligation, but before hypoxia in male neonatal rats. The objective of the current study was to investigate the neuroprotective effects of treatment with IAIPs given immediately or 6 h after HI in both male and female neonatal rats. HI was induced with the Rice-Vannucci method in postnatal (P) day 7 rats. After ligation of the right common carotid artery, P7 rats were exposed to 90 min of hypoxia (8% oxygen). Human plasma-derived IAIPs or placebo (phosphate buffered saline) was given at zero, 24, and 48 h after HI. Brains were perfused, weighed and fixed 72 h after HI at P10. In a second, delayed treatment group, the same procedure was followed except that IAIPs or placebo were given at 6, 24 and 48 h after HI. Separate sham-operated, placebo-treated groups were exposed to identical protocols but were not exposed to carotid artery ligation and remained in room air. Rat sex was recorded. The effects of IAIPs on HI brain injury were examined using histopathological scoring and immunohistochemical analyses of the brain and by using infarct volume measurements on frozen tissue of the entire brain hemispheres ipsilateral and contralateral to HI injury. IAIPs given immediately after HI improved (P < 0.050) histopathological brain injury across and within the cingulate, caudate/putamen, thalamus, hippocampus and parietal cortex in males, but not in females. In contrast, IAIPs given immediately after HI reduced (P < 0.050) infarct volumes of the hemispheres ipsilateral to HI injury in similarly both the males and females. Treatment with IAIPs also resulted in higher (P < 0.050) brain weights compared with the placebo-treated HI group, reduced (P < 0.050) neuronal and non-neuronal cell death in the cortex and total hemisphere, and also increased the total area of oligodendrocytes determined by CNPase in the ipsilateral hemisphere and corpus callosum (P < 0.050) of male, but not female subjects exposed to HI. Delayed treatment with IAIPs 6 h after HI did not improve histopathological brain injury in males or females, but resulted in higher (P < 0.050) brain weights compared with the placebo-treated HI males. Therefore, treatment with IAIPs immediately after HI improved brain weights and reduced neuropathological brain injury and cell death in male rats, and reduced infarct volume in both male and female neonatal rats. We conclude that IAIPs exert neuroprotective effects after exposure to HI in neonatal rats and may exhibit some sex-related differential effects.


Subject(s)
Alpha-Globulins/pharmacology , Hypoxia-Ischemia, Brain/pathology , Neuroprotective Agents/pharmacology , Animals , Animals, Newborn , Brain/drug effects , Brain/pathology , Female , Humans , Male , Rats , Rats, Wistar
10.
Antioxid Redox Signal ; 30(14): 1746-1759, 2019 05 10.
Article in English | MEDLINE | ID: mdl-29943622

ABSTRACT

AIMS: Peptide receptor radionuclide therapy (PRRT) is in clinical use today to treat metastatic neuroendocrine tumors. Infused, radiolabeled, somatostatin analog peptides target tumors that are killed by irradiation damage. The peptides, however, are also retained in kidneys due to glomerular filtration, and the administered doses must be limited to avoid kidney damage. The human radical scavenger and antioxidant, α1-microglobulin (A1M), has previously been shown to protect bystander tissue against irradiation damage and has pharmacokinetic and biodistribution properties similar to somatostatin analogs. In this study, we have investigated if A1M can be used as a renal protective agent in PRRT. RESULTS: We describe nephroprotective effects of human recombinant A1M on the short- and long-term renal damage observed following lutetium 177 (177Lu)-DOTATATE (150 MBq) exposure in BALB/c mice. After 1, 4, and 8 days (short term), 177Lu-DOTATATE injections resulted in increased formation of DNA double-strand breaks in the renal cortex, upregulated expression of apoptosis and stress response-related genes, and proteinuria (albumin in urine), all of which were significantly suppressed by coadministration of A1M (7 mg/kg). After 6, 12, and 24 weeks (long term), 177Lu-DOTATATE injections resulted in increased animal death, kidney lesions, glomerular loss, upregulation of stress genes, proteinuria, and plasma markers of reduced kidney function, all of which were suppressed by coadministration of A1M. Innovation and Conclusion: This study demonstrates that A1M effectively inhibits radiation-induced renal damage. The findings suggest that A1M may be used as a radioprotector during clinical PRRT, potentially facilitating improved tumor control and enabling more patients to receive treatment.


Subject(s)
Alpha-Globulins/pharmacology , Antioxidants/pharmacology , Kidney/drug effects , Kidney/radiation effects , Octreotide/analogs & derivatives , Organometallic Compounds/administration & dosage , Radiation-Protective Agents/pharmacology , Animals , Biomarkers , Gene Expression Profiling , Histones/metabolism , Humans , Kidney/pathology , Mice , Models, Animal , Octreotide/administration & dosage , Survival Rate , Time Factors
11.
Exp Eye Res ; 173: 13-23, 2018 08.
Article in English | MEDLINE | ID: mdl-29678721

ABSTRACT

The purpose of this study was to explore acute tissue reactions, ultrastructural photoreceptor morphology with emphasis on inner segments, and the effect of antioxidant treatment in an in vitro model of rhegmatogenous retinal detachment (RRD). A previously described method of RRD simulation was used with adult retinal porcine explants kept free-floating in culture medium with or without treatment with the radical scavenger α1-microglobulin (A1M). Explants were examined at 5 time points from 1 to 24 h using transmission electron microscopy as well as quantitative real-time PCR (RT-PCR) to quantify gene expression of the cell stress marker heat shock protein 70 (Hsp70) and oxidative stress marker heme oxygenase (HO-1). The culture medium level of the cell damage marker lactate dehydrogenase (LDH) and oxidative stress DNA damage marker 8-Oxo-2'-deoxyguanosine (8-OHdG) was also assessed at each time point. We found that the levels of Hsp70 and LDH rapidly increased in both groups, and at 3 and 6 h, Hsp70 was significantly higher in A1M treated retinas. At 24 h, Hsp70 and LDH, as well as 8-OHdG were significantly lower compared with controls, whereas the tissue level of HO-1 was significantly higher. Progressive ultrastructural photoreceptor changes were seen in untreated control explants from 1 h and onwards including outer segment shortening and loss, disruption of organelles within the inner segments and loss of perikarya in the outer nuclear layer. Inner segment pathology was more rapid and extensive in rods compared with in cones. In A1M treated counterparts, damage to rod inner segment mitochondria was significantly higher after 1 h of culture, but after this time, no statistical difference was found. At 24 h, cone inner segment mitochondrial disruption was significantly higher in control retinas and the number of surviving perikarya lower. From our results, we conclude that retinal explants elicit acute cell stress reactions when placed in culture without physical support simulating a detached retina floating in the vitreous space. Photoreceptors rapidly display degenerative changes including extensive damage to inner segment mitochondria indicating loss of energy transduction as an early key event. A1M increases initial mitochondrial stress in the rods, however, subsequent pathology is attenuated by the treatment, highlighting the dynamics of protective as well as disruptive oxidative stress reactions in the detached retina.


Subject(s)
Acute-Phase Reaction/etiology , Alpha-Globulins/pharmacology , Protease Inhibitors/pharmacology , Retinal Detachment/drug therapy , Retinal Photoreceptor Cell Inner Segment/ultrastructure , 8-Hydroxy-2'-Deoxyguanosine , Acute-Phase Reaction/genetics , Acute-Phase Reaction/pathology , Animals , Antioxidants/pharmacology , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Free Radical Scavengers/pharmacology , Gene Expression Regulation/physiology , HSP70 Heat-Shock Proteins/genetics , Heme Oxygenase-1/genetics , L-Lactate Dehydrogenase/metabolism , Microscopy, Electron, Transmission , Oxidative Stress , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Retinal Detachment/genetics , Retinal Detachment/pathology , Swine
12.
Drug Discov Today ; 22(4): 736-743, 2017 04.
Article in English | MEDLINE | ID: mdl-27988357

ABSTRACT

Preeclampsia is a serious pregnancy-specific condition, affecting 10 million women annually worldwide. No specific treatment is currently available. Recent studies have demonstrated abnormal production and accumulation of free fetal hemoglobin in the preeclamptic placenta, and identified subsequent leakage into the maternal circulation as an important factor in the development of preeclampsia. A recombinant version of alpha-1-microglobulin, an endogenous well-characterized heme and radical scavenger, has been developed. Intravenous administration of recombinant alpha-1-microglobulin in animal models has been proved to eliminate or significantly reduce the manifestations of preeclampsia. Recombinant alpha-1-microglobulin has the potential to become the first specific therapy for preeclampsia.


Subject(s)
Alpha-Globulins/pharmacology , Alpha-Globulins/therapeutic use , Pre-Eclampsia/drug therapy , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Animals , Female , Humans , Placenta/drug effects , Pregnancy
13.
J Biol Chem ; 291(26): 13789-801, 2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27143355

ABSTRACT

Fibroblasts are central to wound healing and fibrosis through TGFß1-triggered differentiation into contractile, α-smooth muscle actin (α-SMA)-positive myofibroblasts. This is mediated by accumulation of a pericellular matrix of hyaluronan (HA) and the HA-dependent co-localization of CD44 with the epidermal growth factor receptor (EGFR). Interactions of HA with hyaladherins, such as inter-α-inhibitor (IαI) and tumor necrosis factor-stimulated gene-6 (TSG-6), are also essential for differentiation. This study investigated the mechanisms involved. TSG-6 and α-SMA had different kinetics of induction by TGFß1, with TSG-6 peaking before α-SMA Si CD44 or EGFR inhibition prevented differentiation but had no effect on TSG-6 expression. TSG-6 was essential for differentiation, and mAb A38 (preventing IαI heavy chain (HC) transfer), HA-oligosaccharides, cobalt, or Si bikunin prevented TSG-6 activity, preventing differentiation. A38 also prevented the EGFR/CD44 association. This suggested that TSG-6/IαI HC interaction was necessary for the effect of TSG-6 and that HC stabilization of HA initiated the CD44/EGFR association. The newly described HC5 was shown to be the principal HC expressed, and its cell surface expression was prevented by siRNA inhibition of TSG-6 or bikunin. HC5 was released by hyaluronidase treatment, confirming its association with cell surface HA. Finally, HC5 knockdown by siRNA confirmed its role in myofibroblast differentiation. The current study describes a novel mechanism linking the TSG-6 transfer of the newly described HC5 to the HA-dependent control of cell phenotype. The interaction of HC5 with cell surface HA was essential for TGFß1-dependent differentiation of fibroblasts to myofibroblasts, highlighting its importance as a novel potential therapeutic target.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Differentiation/physiology , Myofibroblasts/metabolism , Proteinase Inhibitory Proteins, Secretory/metabolism , Transforming Growth Factor beta1/metabolism , Actins/genetics , Actins/metabolism , Alpha-Globulins/pharmacology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Cell Adhesion Molecules/genetics , Cell Differentiation/drug effects , Cell Line , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation/drug effects , Humans , Hyaluronan Receptors/pharmacology , Myofibroblasts/cytology , Transforming Growth Factor beta1/genetics
14.
Am J Physiol Renal Physiol ; 311(3): F640-51, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27170684

ABSTRACT

α1-Microglobulin (A1M) is a low-molecular-weight heme-binding antioxidant protein that is readily filtered by the glomerulus and reabsorbed by proximal tubules. Given these properties, recombinant A1M (rA1M) has been proposed as a renal antioxidant and therapeutic agent. However, little direct evidence to support this hypothesis exists. Hence, we have sought "proof of concept" in this regard. Cultured proximal tubule (HK-2) cells or isolated mouse proximal tubule segments were challenged with a variety of prooxidant insults: 1) hemin, 2) myoglobin; 3) "catalytic" iron, 4) H2O2/Fenton reagents, 5) a Ca(2+) ionophore, 6) antimycin A, or 7) hypoxia (with or without rA1M treatment). HK-2 injury was gauged by the percent lactate dehydrogenase release and 4,5-(dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide uptake. In vivo protection was sought in rA1M-treated mice subjected to 1) graded myohemoglobinura (2, 4, 8, or 9 ml/kg glycerol injection), 2) purified myoglobinemia/uria, or 3) endotoxemia. In vivo injury was assessed by blood urea nitrogen, creatinine, and the expression of redox-sensitive genes (heme oxygenase-1, neutrophil gelatinase-associated lipocalin, and monocyte chemoattractant protein-1 mRNAs). Although rA1M totally blocked in vitro hemin toxicity, equimolar albumin (another heme binder) or 10% serum induced equal protection. rA1M failed to mitigate any nonhemin forms of either in vitro or in vivo injury. A1M appeared to be rapidly degraded within proximal tubules (by Western blot analysis). Surprisingly, rA1M exerted select injury-promoting effects (increased in vitro catalytic iron/antimycin toxicities and increased in vivo monocyte chemoattractant protein-1/neutrophil gelatinase-associated lipocalin mRNA expression after glycerol or endotoxin injection). We conclude that rA1M has questionable utility as a renal antioxidant/cytoprotective agent, particularly in the presence of larger amounts of competitive free heme (e.g., albumin) binders.


Subject(s)
Acute Kidney Injury/prevention & control , Alpha-Globulins/pharmacology , Antioxidants/pharmacology , Kidney Tubules, Proximal/drug effects , Oxidative Stress/drug effects , Recombinant Proteins/pharmacology , Acute Kidney Injury/metabolism , Animals , Antimycin A/pharmacology , Cell Line , Hemin/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Iron/pharmacology , Kidney Tubules, Proximal/metabolism , Mice , Myoglobin/pharmacology , Protective Agents/pharmacology
15.
FEBS J ; 283(9): 1734-47, 2016 05.
Article in English | MEDLINE | ID: mdl-26936634

ABSTRACT

Granzymes are a family of serine proteases that were once thought to function exclusively as mediators of cytotoxic lymphocyte-induced target cell death. However, non-apoptotic roles for granzymes, including granzyme K (GzK), have been proposed. As recent studies have observed elevated levels of GzK in the plasma of patients diagnosed with clinical sepsis, we hypothesized that extracellular GzK induces a proinflammatory response in endothelial cells. In the present study, extracellular GzK proteolytically activated protease-activated receptor-1 leading to increased interleukin 6 and monocyte chemotactic protein 1 production in endothelial cells. Enhanced expression of intercellular adhesion molecule 1 along with an increased capacity for adherence of THP-1 cells was also observed. Characterization of downstream pathways implicated the mitogen-activated protein kinase p38 pathway for intercellular adhesion molecule 1 expression, and both the p38 and the extracellular signal-regulated protein kinases 1 and 2 pathways in cytokine production. GzK also increased tumour necrosis factor α-induced inflammatory adhesion molecule expression. Furthermore, the physiological inhibitor of GzK, inter-α-inhibitor protein, significantly inhibited GzK activity in vitro. In summary, extracellular GzK promotes a proinflammatory response in endothelial cells.


Subject(s)
Granzymes/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Monocytes/drug effects , Receptor, PAR-1/genetics , Alpha-Globulins/pharmacology , Butadienes/pharmacology , Cell Adhesion , Cell Line , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Gene Expression Regulation , Granzymes/antagonists & inhibitors , Granzymes/genetics , Granzymes/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Imidazoles/pharmacology , Interleukin-6/genetics , Interleukin-6/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Monocytes/cytology , Monocytes/metabolism , Nitriles/pharmacology , Proteolysis/drug effects , Pyridines/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-1/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Behav Brain Res ; 302: 88-99, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26778784

ABSTRACT

Neonatal cerebral hypoxia-ischemia (HI) commonly results in cognitive and sensory impairments. Early behavioral experience has been suggested to improve cognitive and sensory outcomes in children and animal models with perinatal neuropathology. In parallel, we previously showed that treatment with immunomodulator Inter-alpha Inhibitor Proteins (IAIPs) improves cellular and behavioral outcomes in neonatal HI injured rats. The purpose of the current study was to evaluate the influences of early experience and typical maturation in combination with IAIPs treatment on spatial working and reference memory after neonatal HI injury. A second aim was to determine the effects of these variables on hippocampal CA1 neuronal morphology. Subjects were divided into two groups that differed with respect to the time when exposed to eight arm radial water maze testing: Group one was tested as juveniles (early experience, Postnatal day (P) 36-61) and adults (P88-113), and Group two was tested in adulthood only (P88-113; without early experience). Three treatment conditions were included in each experience group (HI+Vehicle, HI+IAIPs, and Sham subjects). Incorrect arm entries (errors) were compared between treatment and experience groups across three error types (reference memory (RM), working memory incorrect (WMI), working memory correct (WMC)). Early experience led to improved working memory performance regardless of treatment. Combining IAIPs intervention with early experience provided a long-term behavioral advantage on the WMI component of the task in HI animals. Anatomically, early experience led to a decrease in the average number of basal dendrites per CA1 pyramidal neuron for IAIP treated subjects and a significant reduction in basal dendritic length in control subjects, highlighting the importance of pruning in typical early life learning. Our results support the hypothesis that early behavioral experience combined with IAIPs improve outcome on a relativity demanding cognitive task, beyond that of a single intervention strategy, and appears to facilitate neuronal plasticity following neonatal brain injury.


Subject(s)
Aging , Alpha-Globulins/pharmacology , Hypoxia-Ischemia, Brain/complications , Memory Disorders , Memory, Short-Term/physiology , Neuronal Plasticity/physiology , Analysis of Variance , Animals , Animals, Newborn , CA1 Region, Hippocampal/pathology , Dendrites/drug effects , Dendrites/pathology , Dendrites/ultrastructure , Disease Models, Animal , Hypoxia-Ischemia, Brain/pathology , Male , Maze Learning/drug effects , Memory Disorders/drug therapy , Memory Disorders/etiology , Memory Disorders/pathology , Memory, Short-Term/drug effects , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/ultrastructure , Rats , Rats, Wistar , Silver Staining
17.
Endocr Regul ; 49(1): 37-45, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25687679

ABSTRACT

It has been shown that following endogenous gonadotropin surge, oocyte-cumulus complexes (OCC) synthesize hyaluronan (HA) in a process called cumulus expansion. During this process, HA associates with proteins and proteoglycans to form the expanded HA-rich oocyte-cumulus extracellular matrix (ECM), where the heavy chains of the serum derived inter-α-trypsin inhibitor family (IαI) bind covalently to HA. No study has been performed on the occurrence and regulation of this process during oocyte maturation in species other than mouse and pig, although, the heavy chains (of IαI)-HA complex was purified from human amniotic membrane. The present review pointing out that: 1/ formation of expanded HA-rich oocyte-cumulus ECM is dependent on the presence of IαI molecules, 2/ the heavy chains of IαI molecules identified in the serum are covalently linked to HA during cumulus expansion in mouse and pig, 3/ the family of IαI molecules can freely cross the blood-follicle barrier, and the follicular fluid collected at any stage of folliculogenesis can be successfully used instead of serum to form expanded cumulus ECM in pig, and 4/ proteins of the IαI family can affect reproductive process by modulating the expression of a large number of cellular genes during a preovulatory period. Finally, this review provides clear evidence that IαI family members present in the serum or follicular fluid become responsible for cumulus expansion, as without these proteins, expanded cumulus HA-rich ECM is not formed and HA is released into medium.


Subject(s)
Alpha-Globulins/physiology , Cumulus Cells/cytology , Cumulus Cells/physiology , Extracellular Matrix/physiology , Follicular Phase/physiology , Ovarian Follicle/cytology , Alpha-Globulins/pharmacology , Animals , Cell Proliferation/drug effects , Female , Follicular Fluid/metabolism , Humans , Mice , Ovarian Follicle/physiology , Swine
18.
Am J Physiol Renal Physiol ; 306(4): F442-8, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24338823

ABSTRACT

Extracellular fetal hemoglobin (HbF) and adult hemoglobin (HbA) are proinflammatory and generate ROS. Increased plasma levels of extracellular HbF have recently been reported to occur in early preeclampsia. α1-Microglobulin (A1M) is a physiological heme-binding protein and radical scavenger that has been shown to counteract vascular permeability increases induced by HbA in the perfused placenta. The present study was performed to investigate whether HbF and HbA will increase glomerular permeability in vivo and to test whether A1M and tempol, a ROS scavenger, can prevent their effects. Anesthetized Wistar rats were continuously infused intravenously with either HbA, HbF, or cyano-inactivated HbF together with FITC-Ficoll-70/400, inulin, and (51)Cr-labeled EDTA for 2 h. Plasma samples and urine samples (left ureter) were taken repeatedly and analyzed by high-performance size exclusion chromatography to assess glomerular sieving coefficients for Ficoll of radius 10-80 Å. In separate experiments, A1M or tempol was given before and during Hb infusions. Extracellular HbF caused rapid, transient increases in glomerular permeability to large Ficoll molecules (50-80Å), contrary to the effects of HbA and cyano-inactivated HbF. For HbF, glomerular sieving coefficients for Ficoll of radius 60Å increased from 3.85 ± 0.85 × 10(-5) to 2.60 ± 0.96 × 10(-4) at 15 min, changes that were abrogated by tempol and reduced by A1M. In conclusion, our data demonstrate that extracellular HbF, infused systemically, can acutely increase glomerular permeability through inducing oxidative stress.


Subject(s)
Alpha-Globulins/pharmacology , Antioxidants/pharmacology , Cyclic N-Oxides/pharmacology , Fetal Hemoglobin/pharmacology , Kidney Glomerulus/drug effects , Animals , Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/physiology , Kidney Glomerulus/metabolism , Male , Permeability/drug effects , Rats , Rats, Wistar , Spin Labels
19.
Curr Opin Obstet Gynecol ; 25(6): 448-55, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24185004

ABSTRACT

PURPOSE OF REVIEW: Preeclampsia, one of the leading causes of pregnancy complications, affects 3-7% of pregnant women. This review summarizes the present knowledge of a new potential cause of the disease and suggests a method for its prediction/diagnosis and a possible treatment, both based on the recent findings on the involvement of fetal hemoglobin (HbF) and the heme and radical scavenging protein A1M (alpha-1-microglobulin). RECENT FINDINGS: Gene and protein profiling studies have independently shown that increased amount of free HbF is accumulated in the preeclampsia placenta. As a result of a predominantly oxidative damage to the blood-placenta barrier, HbF leaks over to the maternal blood circulation. Elevated levels can be measured already in the first trimester, and later in pregnancy, the levels correlate with the blood pressure in women with preeclampsia. Ex-vivo data show that the human protein A1M, an endogeneous antioxidation protection protein, can prevent Hb-induced damage to the placenta, restore the blood-placental barrier and prevent maternal tissue damage. SUMMARY: Free HbF may provide both a predictive and a diagnostic clinical biomarker from the first trimester. A1M has the potential as a future pharmacological treatment for preeclampsia.


Subject(s)
Alpha-Globulins/metabolism , Antioxidants/metabolism , Fetal Hemoglobin/metabolism , Oxidative Stress , Placenta/metabolism , Pre-Eclampsia/etiology , Alpha-Globulins/pharmacology , Antioxidants/pharmacology , Biomarkers/metabolism , Female , Fetal Hemoglobin/pharmacology , Gene Expression Profiling , Humans , Placenta/physiopathology , Placental Circulation , Pre-Eclampsia/diagnosis , Pre-Eclampsia/drug therapy , Pre-Eclampsia/metabolism , Pregnancy
20.
R I Med J (2013) ; 96(2): 16-8, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23641420

ABSTRACT

ProThera Biologics is a development stage bio-therapeutics company in East Providence, Rhode Island. The company was founded in 2002 to focus on the critical role and commercial potential of Inter-alpha Inhibitor Proteins (IAIP) for treating acute life-threatening inflammatory diseases. The discovery research originated in the basic research laboratories of the co-founders, Yow-Pin Lim, MD, PhD, and Douglas C. Hixson, PhD, at Rhode Island Hospital, a Lifespan partner. The company is backed by the Slater Technology Fund and has received research grants from the Rhode Island State Science and Technology Council (RI STAC) as well as continuous funding from the National Institutes of Health (NIH), with several Phase I and II Small Business Innovation Research (SBIR) grants over the past 10 years. ProThera has developed a novel process to purify Inter-alpha Inhibitor Proteins from source material, and has conducted groundbreaking research into the usage of IAIP to fight systemic inflammation.


Subject(s)
Alpha-Globulins/pharmacology , Anthrax/drug therapy , Anti-Infective Agents/pharmacology , Bacillus anthracis/drug effects , Biomedical Research , National Institutes of Health (U.S.) , Sepsis/drug therapy , Adjuvants, Immunologic , Alpha-Globulins/analysis , Animals , Biomarkers , Drug Discovery , Humans , Research Personnel , Rhode Island , United States
SELECTION OF CITATIONS
SEARCH DETAIL
...