Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
J Headache Pain ; 18(1): 16, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28176234

ABSTRACT

BACKGROUND: Frequent mild head injuries or concussion along with the presence of headache may contribute to the persistence of concussion symptoms. METHODS: In this study, the acute effects of recovery between mild head injuries and the frequency of injuries on a headache behavior, trigeminal allodynia, was assessed using von Frey testing up to one week after injury, while histopathological changes in the trigeminal pain pathway were evaluated using western blot, ELISA and immunohistochemistry.  RESULTS: A decreased recovery time combined with an increased mild closed head injury (CHI) frequency results in reduced trigeminal allodynia thresholds compared to controls. The repetitive CHI group with the highest injury frequency showed the greatest reduction in trigeminal thresholds along with greatest increased levels of calcitonin gene-related peptide (CGRP) in the trigeminal nucleus caudalis. Repetitive CHI resulted in astrogliosis in the central trigeminal system, increased GFAP protein levels in the sensory barrel cortex, and an increased number of microglia cells in the trigeminal nucleus caudalis. CONCLUSIONS: Headache behavior in rats is dependent on the injury frequency and recovery interval between mild head injuries. A worsening of headache behavior after repetitive mild head injuries was concomitant with increases in CGRP levels, the presence of astrocytosis, and microglia proliferation in the central trigeminal pathway. Signaling between neurons and proliferating microglia in the trigeminal pain system may contribute to the initiation of acute headache after concussion or other traumatic brain injuries.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Craniocerebral Trauma/complications , Gliosis/etiology , Headache/etiology , Hyperalgesia/etiology , Microglia/metabolism , Trigeminal Caudal Nucleus/metabolism , Trigeminal Nerve Diseases/etiology , Animals , Disease Models, Animal , Headache/metabolism , Hyperalgesia/metabolism , Male , Rats , Rats, Sprague-Dawley , Trigeminal Nerve Diseases/metabolism
2.
Sci Adv ; 8(11): eabj4716, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35302860

ABSTRACT

Dysregulation of mitochondrial Ca2+ homeostasis has been linked to neurodegenerative diseases. Mitochondrial Ca2+ uptake is mediated via the calcium uniporter complex that is primarily regulated by MICU1, a Ca2+-sensing gatekeeper. Recently, human patients with MICU1 loss-of-function mutations were diagnosed with neuromuscular and cognitive impairments. While studies in patient-derived cells revealed altered mitochondrial calcium signaling, the neuronal pathogenesis was difficult to study. To fill this void, we created a neuron-specific MICU1-KO mouse model. These animals show progressive, abnormal motor and cognitive phenotypes likely caused by the degeneration of motor neurons in the spinal cord and the cortex. We found increased susceptibility to mitochondrial Ca2+ overload-induced excitotoxic insults and cell death in MICU1-KO neurons and MICU1-deficient patient-derived cells, which can be blunted by inhibiting the mitochondrial permeability transition pore. Thus, our study identifies altered neuronal mitochondrial Ca2+ homeostasis as causative in the clinical symptoms of MICU1-deficient patients and highlights potential therapeutic targets.


Subject(s)
Cation Transport Proteins , Mitochondrial Membrane Transport Proteins , Neurodegenerative Diseases , Animals , Calcium/metabolism , Calcium Signaling , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Humans , Mice , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology
3.
J Neurotrauma ; 35(18): 2195-2207, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29471717

ABSTRACT

Populations of neural stem cells (NSCs) reside in a number of defined niches in the adult central nervous system (CNS) where they continually give rise to mature cell types throughout life, including newly born neurons. In addition to the prototypical niches of the subventricular zone (SVZ) and subgranular zone (SGZ) of the hippocampal dentate gyrus, novel stem cell niches that are also neurogenic have recently been identified in multiple midline structures, including circumventricular organs (CVOs) of the brain. These resident NSCs serve as a homeostatic source of new neurons and glial cells under intact physiological conditions. Importantly, they may also have the potential for reparative processes in pathological states such as traumatic spinal cord injury (SCI) and traumatic brain injury (TBI). As the response in these novel CVO stem cell niches has been characterized after stroke but not following SCI or TBI, we quantitatively assessed cell proliferation and the neuronal and glial lineage fate of resident NSCs in three CVO nuclei-area postrema (AP), median eminence (ME), and subfornical organ (SFO) -in rat models of cervical contusion-type SCI and controlled cortical impact (CCI)-induced TBI. Using bromodeoxyuridine (BrdU) labeling of proliferating cells, we find that TBI significantly enhanced proliferation in AP, ME, and SFO, whereas cervical SCI had no effects at early or chronic time-points post-injury. In addition, SCI did not alter NSC differentiation profile into doublecortin-positive neuroblasts, GFAP-expressing astrocytes, or Olig2-labeled cells of the oligodendrocyte lineage within AP, ME, or SFO at both time-points. In contrast, CCI induced a pronounced increase in Sox2- and doublecortin-labeled cells in the AP and Iba1-labeled microglia in the SFO. Lastly, plasma derived from CCI animals significantly increased NSC expansion in an in vitro neurosphere assay, whereas plasma from SCI animals did not exert such an effect, suggesting that signaling factors present in blood may be relevant to stimulating CVO niches after CNS injury and may explain the differential in vivo effects of SCI and TBI on the novel stem cell niches.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Circumventricular Organs/cytology , Neural Stem Cells/physiology , Spinal Cord Injuries/physiopathology , Stem Cell Niche , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Cervical Cord , Doublecortin Protein , Female , Neurogenesis/physiology , Rats , Rats, Sprague-Dawley
4.
J Neurotrauma ; 33(8): 748-60, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26472135

ABSTRACT

The pain-signaling molecules, nitric oxide synthase (NOS) and calcitonin gene-related peptide (CGRP), are implicated in the pathophysiology of post-traumatic headache (PTH) as they are for migraine. This study assessed the changes of inducible NOS (iNOS) and its cellular source in the trigeminal pain circuit, as well as the relationship between iNOS and CGRP after controlled cortical impact (CCI) injury in mice. The effects of a CGRP antagonist (MK8825) and sumatriptan on iNOS messenger RNA (mRNA) and protein were compared to vehicle at 2 weeks postinjury. Changes in CGRP levels in the trigeminal nucleus caudalis (TNC) in iNOS knockouts with CCI were compared to wild-type (WT) mice at 3 days and 2 weeks post injury. Trigeminal allodynia and photosensitivity were measured. MK8825 and sumatriptan increased allodynic thresholds in CCI groups compared to vehicle (p < 0.01), whereas iNOS knockouts were not different from WT. Photosensitivity was attenuated in MK8825 mice and iNOS knockouts compared to WT (p < 0.05). MK8825 and sumatriptan reduced levels of iNOS mRNA and iNOS immunoreactivity in the TNC and ganglia (p < 0.01). Differences in iNOS cellular localization were found between the trigeminal ganglia and TNC. Although the knockout of iNOS attenuated CGRP at 3 days (p < 0.05), it did not reduce CGRP at 2 weeks. CGRP immunoreactivity was found in the meningeal layers post-CCI, while negligible in controls. Findings support the importance of interactions between CGRP and iNOS in mediating allodynia, as well as the individual roles in photosensitivity. Mitigating prolonged increases in CGRP may be a promising intervention for treating acute PTH.


Subject(s)
Brain Injuries, Traumatic/metabolism , Calcitonin Gene-Related Peptide/metabolism , Disease Models, Animal , Hyperalgesia/metabolism , Nitric Oxide Synthase Type II/deficiency , Photosensitivity Disorders/metabolism , Animals , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/genetics , Calcitonin Gene-Related Peptide/antagonists & inhibitors , Calcitonin Gene-Related Peptide/genetics , Hyperalgesia/drug therapy , Hyperalgesia/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Photic Stimulation/methods , Photosensitivity Disorders/drug therapy , Photosensitivity Disorders/genetics , Pyridines/pharmacology , Pyridines/therapeutic use , Random Allocation , Spiro Compounds/pharmacology , Spiro Compounds/therapeutic use , Sumatriptan/pharmacology , Sumatriptan/therapeutic use , Trigeminal Nerve/metabolism , Trigeminal Nerve/pathology
SELECTION OF CITATIONS
SEARCH DETAIL