1
|
Balcomb K, Johnston C, Kavanagh T, Leitner D, Schneider J, Halliday G, Wisniewski T, Sunde M, Drummond E. SMOC1 colocalizes with Alzheimer's disease neuropathology and delays Aβ aggregation. Acta Neuropathol 2024; 148:72. [PMID: 39585417 PMCID: PMC11588930 DOI: 10.1007/s00401-024-02819-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 11/26/2024]
Abstract
SMOC1 has emerged as one of the most significant and consistent new biomarkers of early Alzheimer's disease (AD). Recent studies show that SMOC1 is one of the earliest changing proteins in AD, with levels in the cerebrospinal fluid increasing many years before symptom onset. Despite this clear association with disease, little is known about the role of SMOC1 in AD or its function in the brain. Therefore, the aim of this study was to examine the distribution of SMOC1 in human AD brain tissue and to determine if SMOC1 influenced amyloid beta (Aβ) aggregation. The distribution of SMOC1 in human brain tissue was assessed in 3 brain regions (temporal cortex, hippocampus, and frontal cortex) using immunohistochemistry in a cohort of 73 cases encompassing advanced AD, mild cognitive impairment (MCI), preclinical AD, and cognitively normal controls. The Aβ- and phosphorylated tau-interaction with SMOC1 was assessed in control, MCI, and advanced AD human brain tissue using co-immunoprecipitation, and the influence of SMOC1 on Aβ aggregation kinetics was assessed using Thioflavin-T assays and electron microscopy. SMOC1 strongly colocalized with a subpopulation of amyloid plaques in AD (43.8 ± 2.4%), MCI (32.8 ± 5.4%), and preclinical AD (28.3 ± 6.4%). SMOC1 levels in the brain strongly correlated with plaque load, irrespective of disease stage. SMOC1 also colocalized with a subpopulation of phosphorylated tau aggregates in AD (9.6 ± 2.6%). Co-immunoprecipitation studies showed that SMOC1 strongly interacted with Aβ in human MCI and AD brain tissue and with phosphorylated tau in human AD brain tissue. Thioflavin-T aggregation assays showed that SMOC1 significantly delayed Aβ aggregation in a dose-dependent manner, and electron microscopy confirmed that the Aβ fibrils generated in the presence of SMOC1 had an altered morphology. Overall, our results emphasize the importance of SMOC1 in the onset and progression of AD and suggest that SMOC1 may influence pathology development in AD.
Collapse
Affiliation(s)
- Kaleah Balcomb
- Brain and Mind Centre and School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Caitlin Johnston
- School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Tomas Kavanagh
- Brain and Mind Centre and School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Dominique Leitner
- Center for Cognitive Neurology, Department of Neurology, Grossman School of Medicine, New York University, New York, NY, 10016, USA
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Julie Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison Street, Suite 1000, Chicago, IL, 60612, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Glenda Halliday
- Brain and Mind Centre and School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Thomas Wisniewski
- Center for Cognitive Neurology, Department of Neurology, Grossman School of Medicine, New York University, New York, NY, 10016, USA
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Margaret Sunde
- School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Eleanor Drummond
- Brain and Mind Centre and School of Medical Sciences, University of Sydney, Camperdown, NSW, 2050, Australia.
| |
Collapse
|
2
|
Zhou H, Hu YY, Tang ZX, Jiang ZB, Huang J, Zhang T, Shen HY, Ye XP, Huang XY, Wang X, Zhou T, Bai XL, Zhu Q, Shi LE. Calcium Transport and Enrichment in Microorganisms: A Review. Foods 2024; 13:3612. [PMID: 39594028 PMCID: PMC11593130 DOI: 10.3390/foods13223612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/31/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
Calcium is a vital trace element for the human body, and its deficiency can result in a range of pathological conditions, including rickets and osteoporosis. Despite the numerous types of calcium supplements currently available on the market, these products are afflicted with a number of inherent deficiencies, such as low calcium content, poor aqueous solubility, and low human absorption rate. Many microorganisms, particularly beneficial microorganisms, including edible fungi, lactic acid bacteria, and yeast, are capable of absorbing and enriching calcium, a phenomenon that has been widely documented. This opens the door to the potential utilization of microorganisms as novel calcium enrichment carriers. However, the investigation of calcium-rich foods from microorganisms still faces many obstacles, including a poor understanding of calcium metabolic pathways in microorganisms, a relatively low calcium enrichment rate, and the slow growth of strains. Therefore, in order to promote the development of calcium-rich products from microorganisms, this paper provides an overview of the impacts of calcium addition on strain growth, calcium enrichment rate, antioxidant system, and secondary metabolite production. Additionally, it highlights calcium transport and enrichment mechanisms in microorganism cells and offers a detailed account of the progress made on calcium-binding proteins, calcium transport pathways, and calcium storage and release. This paper offers insights for further research on the relevant calcium enrichment in microorganism cells.
Collapse
Affiliation(s)
- Hai Zhou
- Department of Biotechnology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (H.Z.); (Y.-Y.H.); (Z.-B.J.); (J.H.); (T.Z.); (H.-Y.S.); (X.-P.Y.); (X.-Y.H.); (X.W.); (T.Z.); (X.-L.B.); (Q.Z.)
| | - Yan-Yu Hu
- Department of Biotechnology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (H.Z.); (Y.-Y.H.); (Z.-B.J.); (J.H.); (T.Z.); (H.-Y.S.); (X.-P.Y.); (X.-Y.H.); (X.W.); (T.Z.); (X.-L.B.); (Q.Z.)
| | - Zhen-Xing Tang
- School of Culinary Art, Tourism College of Zhejiang, Hangzhou 311231, China
| | - Zhong-Bao Jiang
- Department of Biotechnology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (H.Z.); (Y.-Y.H.); (Z.-B.J.); (J.H.); (T.Z.); (H.-Y.S.); (X.-P.Y.); (X.-Y.H.); (X.W.); (T.Z.); (X.-L.B.); (Q.Z.)
| | - Jie Huang
- Department of Biotechnology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (H.Z.); (Y.-Y.H.); (Z.-B.J.); (J.H.); (T.Z.); (H.-Y.S.); (X.-P.Y.); (X.-Y.H.); (X.W.); (T.Z.); (X.-L.B.); (Q.Z.)
| | - Tian Zhang
- Department of Biotechnology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (H.Z.); (Y.-Y.H.); (Z.-B.J.); (J.H.); (T.Z.); (H.-Y.S.); (X.-P.Y.); (X.-Y.H.); (X.W.); (T.Z.); (X.-L.B.); (Q.Z.)
| | - Hui-Yang Shen
- Department of Biotechnology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (H.Z.); (Y.-Y.H.); (Z.-B.J.); (J.H.); (T.Z.); (H.-Y.S.); (X.-P.Y.); (X.-Y.H.); (X.W.); (T.Z.); (X.-L.B.); (Q.Z.)
| | - Xin-Pei Ye
- Department of Biotechnology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (H.Z.); (Y.-Y.H.); (Z.-B.J.); (J.H.); (T.Z.); (H.-Y.S.); (X.-P.Y.); (X.-Y.H.); (X.W.); (T.Z.); (X.-L.B.); (Q.Z.)
| | - Xuan-Ya Huang
- Department of Biotechnology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (H.Z.); (Y.-Y.H.); (Z.-B.J.); (J.H.); (T.Z.); (H.-Y.S.); (X.-P.Y.); (X.-Y.H.); (X.W.); (T.Z.); (X.-L.B.); (Q.Z.)
| | - Xiang Wang
- Department of Biotechnology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (H.Z.); (Y.-Y.H.); (Z.-B.J.); (J.H.); (T.Z.); (H.-Y.S.); (X.-P.Y.); (X.-Y.H.); (X.W.); (T.Z.); (X.-L.B.); (Q.Z.)
| | - Ting Zhou
- Department of Biotechnology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (H.Z.); (Y.-Y.H.); (Z.-B.J.); (J.H.); (T.Z.); (H.-Y.S.); (X.-P.Y.); (X.-Y.H.); (X.W.); (T.Z.); (X.-L.B.); (Q.Z.)
| | - Xue-Lian Bai
- Department of Biotechnology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (H.Z.); (Y.-Y.H.); (Z.-B.J.); (J.H.); (T.Z.); (H.-Y.S.); (X.-P.Y.); (X.-Y.H.); (X.W.); (T.Z.); (X.-L.B.); (Q.Z.)
| | - Qin Zhu
- Department of Biotechnology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (H.Z.); (Y.-Y.H.); (Z.-B.J.); (J.H.); (T.Z.); (H.-Y.S.); (X.-P.Y.); (X.-Y.H.); (X.W.); (T.Z.); (X.-L.B.); (Q.Z.)
| | - Lu-E Shi
- Department of Biotechnology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (H.Z.); (Y.-Y.H.); (Z.-B.J.); (J.H.); (T.Z.); (H.-Y.S.); (X.-P.Y.); (X.-Y.H.); (X.W.); (T.Z.); (X.-L.B.); (Q.Z.)
| |
Collapse
|
3
|
Balcomb K, Johnston C, Kavanagh T, Leitner D, Schneider J, Halliday G, Wisniewski T, Sunde M, Drummond E. SMOC1 colocalizes with Alzheimer's disease neuropathology and delays Aβ aggregation. RESEARCH SQUARE 2024:rs.3.rs-5229472. [PMID: 39574902 PMCID: PMC11581049 DOI: 10.21203/rs.3.rs-5229472/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
SMOC1 has emerged as one of the most significant and consistent new biomarkers of early Alzheimer's disease (AD). Recent studies show that SMOC1 is one of the earliest changing proteins in AD, with levels in the cerebrospinal fluid increasing many years before symptom onset. Despite this clear association with disease, little is known about the role of SMOC1 in AD or its function in the brain. Therefore, the aim of this study was to examine the distribution of SMOC1 in human AD brain tissue and to determine if SMOC1 influenced amyloid beta (Aβ) aggregation. The distribution of SMOC1 in human brain tissue was assessed in 3 brain regions (temporal cortex, hippocampus, frontal cortex) using immunohistochemistry in a cohort of 73 cases encompassing advanced AD, mild cognitive impairment (MCI), preclinical AD and cognitively normal controls. The Aβ- and phosphorylated tau-interaction with SMOC1 was assessed in control, MCI and advanced AD human brain tissue using co-immunoprecipitation, and the influence of SMOC1 on Aβ aggregation kinetics was assessed using Thioflavin T assays and electron microscopy. SMOC1 strongly colocalized with a subpopulation of amyloid plaques in AD (43.8±2.4%), MCI (32.8±5.4%) and preclinical AD (28.3±6.4%). SMOC1 levels in the brain strongly correlated with plaque load, irrespective of disease stage. SMOC1 also colocalized with a subpopulation of phosphorylated tau aggregates in AD (9.6±2.6%). Co-immunoprecipitation studies showed that SMOC1 strongly interacted with Aβ in human MCI and AD brain tissue and with phosphorylated tau in human AD brain tissue. Thioflavin T aggregation assays showed that SMOC1 significantly delayed Aβ aggregation in a dose-dependent manner, and electron microscopy confirmed that the Aβ fibrils generated in the presence of SMOC1 had an altered morphology. Overall, our results emphasize the importance of SMOC1 in the onset and progression of AD and suggest that SMOC1 may influence pathology development in AD.
Collapse
|
4
|
Vanessa Becerra-Hernández L, Casanova MF, Buriticá E. Cortical calretinin-positive neurons: Functional and ontogenetic characteristics and their relationship to brain pathologies. Brain Res 2024; 1846:149285. [PMID: 39490954 DOI: 10.1016/j.brainres.2024.149285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 06/02/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024]
Abstract
Cortical GABAergic interneurons can be classified according to electrophysiological, biochemical, and/or morphological criteria. In humans, the use of calcium-binding proteins allows us to differentiate three subpopulations of GABAergic interneurons with minimal overlap. Cortical calretinin-positive neurons mainly include bipolar and double-bouquet morphologies, with a largely non-rapid and adaptive firing pattern, originating from the ganglionic eminence and the ventricular and subventricular regions of the developing brain. These cells are distributed from layer I to VI of the neocortex, with predominance in layers II and III. Given their morphology, distribution of processes, and elucidated synaptic contacts, these neurons are considered important in the control of intraminicolumnar processing through vertical inhibition. They have been extensively studied in the context of pathologies characterized by excitation/inhibition imbalance, such as Alzheimer's disease, epilepsy, traumatic brain injury, and autism. In light of the current evidence, this review considers these aspects in depth and discusses the pathophysiological role and selective vulnerability (pathoclisis) vs. the resistance that these interneurons can present against different types of injury.
Collapse
Affiliation(s)
- Lina Vanessa Becerra-Hernández
- Centro de Estudios Cerebrales, Facultad de Salud, Universidad del Valle, Cali, Colombia; Departamento de Ciencias Básicas de la Salud, Pontificia Universidad Javeriana, Cali, Colombia.
| | - Manuel F Casanova
- Department of Biomedical Sciences, School of Medicine Greenville, University of South Carolina, Greenville, SC, United States
| | - Efraín Buriticá
- Centro de Estudios Cerebrales, Facultad de Salud, Universidad del Valle, Cali, Colombia
| |
Collapse
|
5
|
Perry JC, Vann SD. Reduction in neurons immunoreactive for calcium-binding proteins in the anteroventral thalamic nuclei of individuals with Down syndrome. Neuroscience 2024; 557:56-66. [PMID: 39127343 DOI: 10.1016/j.neuroscience.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/26/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
The anterior thalamic nuclei are important for cognition, and memory in particular. However, little is known about how the anterior thalamic nuclei are affected in many neurological disorders partly due to difficulties in selective segmentation in in vivo scans, due to their size and location. Post-mortem studies, therefore, remain a valuable source of information about the status of the anterior thalamic nuclei. We used post-mortem tissue to assess the status of the anteroventral thalamic nucleus in Down syndrome using samples from males and females ranging from 22-65 years in age and comparing to tissue from age matched controls. As expected, there was increased beta-amyloid plaque expression in the Down syndrome group. While there was a significant increase in neuronal density in the Down syndrome group, the values showed more variation consistent with a heterogeneous population. The surface area of the anteroventral thalamic nucleus was smaller in the Down syndrome group suggesting the increased neuronal density was due to greater neuronal packing but likely fewer overall neurons. There was a marked reduction in the proportion of neurons immunoreactive for the calcium-binding proteins calbindin, calretinin, and parvalbumin in individuals with Down syndrome. These findings highlight the vulnerability of calcium-binding proteins in the anteroventral nucleus in Down syndrome, which could both be driven by, and exacerbate, Alzheimer-related pathology in this region.
Collapse
Affiliation(s)
- James C Perry
- School of Psychology & Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK
| | - Seralynne D Vann
- School of Psychology & Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK.
| |
Collapse
|
6
|
Geigenmüller JN, Tari AR, Wisloff U, Walker TL. The relationship between adult hippocampal neurogenesis and cognitive impairment in Alzheimer's disease. Alzheimers Dement 2024; 20:7369-7383. [PMID: 39166771 PMCID: PMC11485317 DOI: 10.1002/alz.14179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/14/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024]
Abstract
Neurogenesis persists throughout adulthood in the hippocampus and contributes to specific cognitive functions. In Alzheimer's disease (AD), the hippocampus is affected by pathology and functional impairment early in the disease. Human AD patients have reduced adult hippocampal neurogenesis (AHN) levels compared to age-matched healthy controls. Similarly, rodent AD models show a decrease in AHN before the onset of the classical hallmarks of AD pathology. Conversely, enhancement of AHN can protect against AD pathology and ameliorate memory deficits in both rodents and humans. Therefore, impaired AHN may be a contributing factor of AD-associated cognitive decline, rather than an effect of it. In this review we outline the regulation and function of AHN in healthy individuals, and highlight the relationship between AHN dysfunction and cognitive impairments in AD. The existence of AHN in humans and its relevance in AD patients will also be discussed, with an outlook toward future research directions. HIGHLIGHTS: Adult hippocampal neurogenesis occurs in the brains of mammals including humans. Adult hippocampal neurogenesis is reduced in Alzheimer's disease in humans and animal models.
Collapse
Affiliation(s)
| | - Atefe R. Tari
- The Cardiac Exercise Research Group at Department of Circulation and Medical ImagingFaculty of Medicine and Health SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
- Department of Neurology and Clinical NeurophysiologySt. Olavs University Hospital, Trondheim University HospitalTrondheimNorway
| | - Ulrik Wisloff
- The Cardiac Exercise Research Group at Department of Circulation and Medical ImagingFaculty of Medicine and Health SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
| | - Tara L. Walker
- Clem Jones Centre for Ageing Dementia ResearchQueensland Brain InstituteThe University of QueenslandBrisbaneAustralia
| |
Collapse
|
7
|
Farris T, González-Ochoa S, Mohammed M, Rajakaruna H, Tonello J, Kanagasabai T, Korolkova O, Shimamoto A, Ivanova A, Shanker A. Loss of Mitochondrial Tusc2/Fus1 Triggers a Brain Pro-Inflammatory Microenvironment and Early Spatial Memory Impairment. Int J Mol Sci 2024; 25:7406. [PMID: 39000512 PMCID: PMC11242373 DOI: 10.3390/ijms25137406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Brain pathological changes impair cognition early in disease etiology. There is an urgent need to understand aging-linked mechanisms of early memory loss to develop therapeutic strategies and prevent the development of cognitive impairment. Tusc2 is a mitochondrial-resident protein regulating Ca2+ fluxes to and from mitochondria impacting overall health. We previously reported that Tusc2-/- female mice develop chronic inflammation and age prematurely, causing age- and sex-dependent spatial memory deficits at 5 months old. Therefore, we investigated Tusc2-dependent mechanisms of memory impairment in 4-month-old mice, comparing changes in resident and brain-infiltrating immune cells. Interestingly, Tusc2-/- female mice demonstrated a pro-inflammatory increase in astrocytes, expression of IFN-γ in CD4+ T cells and Granzyme-B in CD8+T cells. We also found fewer FOXP3+ T-regulatory cells and Ly49G+ NK and Ly49G+ NKT cells in female Tusc2-/- brains, suggesting a dampened anti-inflammatory response. Moreover, Tusc2-/- hippocampi exhibited Tusc2- and sex-specific protein changes associated with brain plasticity, including mTOR activation, and Calbindin and CamKII dysregulation affecting intracellular Ca2+ dynamics. Overall, the data suggest that dysregulation of Ca2+-dependent processes and a heightened pro-inflammatory brain microenvironment in Tusc2-/- mice could underlie cognitive impairment. Thus, strategies to modulate the mitochondrial Tusc2- and Ca2+- signaling pathways in the brain should be explored to improve cognitive health.
Collapse
Affiliation(s)
- Tonie Farris
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.F.); (M.M.); (T.K.)
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Salvador González-Ochoa
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Muna Mohammed
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.F.); (M.M.); (T.K.)
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Harshana Rajakaruna
- The Office for Research and Innovation, Meharry Medical College, Nashville, TN 37208, USA;
| | - Jane Tonello
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Thanigaivelan Kanagasabai
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.F.); (M.M.); (T.K.)
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Olga Korolkova
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Akiko Shimamoto
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Alla Ivanova
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.F.); (M.M.); (T.K.)
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Anil Shanker
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
- The Office for Research and Innovation, Meharry Medical College, Nashville, TN 37208, USA;
| |
Collapse
|
8
|
Maiorov SA, Kairat BK, Berezhnov AV, Zinchenko VP, Gaidin SG, Kosenkov AM. Peculiarities of ion homeostasis in neurons containing calcium-permeable AMPA receptors. Arch Biochem Biophys 2024; 754:109951. [PMID: 38452968 DOI: 10.1016/j.abb.2024.109951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/16/2024] [Accepted: 02/28/2024] [Indexed: 03/09/2024]
Abstract
Glutamate excitotoxicity accompanies numerous brain pathologies, including traumatic brain injury, ischemic stroke, and epilepsy. Disturbances of the ion homeostasis, mitochondria dysfunction, and further cell death are considered the main detrimental consequences of excitotoxicity. It is well known that neurons demonstrate different vulnerability to pathological exposures. In this regard, neurons containing calcium-permeable AMPA receptors (CP-AMPARs) may show higher susceptibility to excitotoxicity due to an additional pathway of Ca2+ influx. Here, we demonstrate that neurons containing CP-AMPARs are characterized by the higher amplitude of the glutamate-induced elevation of intracellular Ca2+ concentration ([Ca2+]i) and slower restoration of [Ca2+]i level compared to non-CP-AMPA neurons. Moreover, we have found that NASPM, an antagonist of CP-AMPARs, significantly decreases the amplitude of the [Ca2+]i elevation induced by glutamate or selective AMPARs agonist, 5-fluorowillardiine. In contrast, the antagonists of NMDARs or KARs affect insignificantly. We have also described some peculiarities of Na+, K+, and H+ intracellular dynamics in neurons containing CP-AMPARs. In particular, the amplitude of [Na+]i elevation was lower compared to non-CP-AMPA neurons, whereas the amplitude of [K+]i decrease was higher. We have shown the significant inverse correlation between [K+]i and [Ca2+]i and between intracellular pH and [Na+]i in CP-AMPARs-containing and non-CP-AMPA neurons upon glutamate excitotoxicity. Our data indicate that CP-AMPARs-mediated Ca2+ influx and slow removal of Ca2+ from the cytosol may underlie the vulnerability of the CP-AMPARs-containing neurons to glutamate excitotoxicity. Further studies of the mechanisms mediating the disturbances in ion homeostasis are crucial for developing new approaches for protecting these neurons at brain pathologies.
Collapse
Affiliation(s)
- Sergei A Maiorov
- Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institute of Cell Biophysics of the Russian Academy of Sciences, 142290, Pushchino, Russia
| | | | - Alexey V Berezhnov
- Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institute of Cell Biophysics of the Russian Academy of Sciences, 142290, Pushchino, Russia
| | - Valery P Zinchenko
- Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institute of Cell Biophysics of the Russian Academy of Sciences, 142290, Pushchino, Russia
| | - Sergei G Gaidin
- Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institute of Cell Biophysics of the Russian Academy of Sciences, 142290, Pushchino, Russia.
| | - Artem M Kosenkov
- Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institute of Cell Biophysics of the Russian Academy of Sciences, 142290, Pushchino, Russia.
| |
Collapse
|
9
|
Acharya M, Singh N, Gupta G, Tambuwala MM, Aljabali AAA, Chellappan DK, Dua K, Goyal R. Vitamin D, Calbindin, and calcium signaling: Unraveling the Alzheimer's connection. Cell Signal 2024; 116:111043. [PMID: 38211841 DOI: 10.1016/j.cellsig.2024.111043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 01/13/2024]
Abstract
Calcium is a ubiquitous second messenger that is indispensable in regulating neurotransmission and memory formation. A precise intracellular calcium level is achieved through the concerted action of calcium channels, and calcium exerts its effect by binding to an array of calcium-binding proteins, including calmodulin (CAM), calcium-calmodulin complex-dependent protein kinase-II (CAMK-II), calbindin (CAL), and calcineurin (CAN). Calbindin orchestrates a plethora of signaling events that regulate synaptic transmission and depolarizing signals. Vitamin D, an endogenous fat-soluble metabolite, is synthesized in the skin upon exposure to ultraviolet B radiation. It modulates calcium signaling by increasing the expression of the calcium-sensing receptor (CaSR), stimulating phospholipase C activity, and regulating the expression of calcium channels such as TRPV6. Vitamin D also modulates the activity of calcium-binding proteins, including CAM and calbindin, and increases their expression. Calbindin, a high-affinity calcium-binding protein, is involved in calcium buffering and transport in neurons. It has been shown to inhibit apoptosis and caspase-3 activity stimulated by presenilin 1 and 2 in AD. Whereas CAM, another calcium-binding protein, is implicated in regulating neurotransmitter release and memory formation by phosphorylating CAN, CAMK-II, and other calcium-regulated proteins. CAMK-II and CAN regulate actin-induced spine shape changes, which are further modulated by CAM. Low levels of both calbindin and vitamin D are attributed to the pathology of Alzheimer's disease. Further research on vitamin D via calbindin-CAMK-II signaling may provide newer insights, revealing novel therapeutic targets and strategies for treatment.
Collapse
Affiliation(s)
- Manish Acharya
- Department of Neuropharmacology, School of Pharmaceutical Sciences, Shoolini University, Himachal Pradesh, India
| | - Nicky Singh
- Department of Neuropharmacology, School of Pharmaceutical Sciences, Shoolini University, Himachal Pradesh, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Jaipur 302017, India
| | - Murtaza M Tambuwala
- Lincoln Medical School, Universities of Nottingham and Lincoln College of Science, Brayford Pool Campus, Lincoln LN6 7TS, UK.
| | - Alaa A A Aljabali
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Yarmouk University, Irbid 21163, Jordan.
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia.
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia.
| | - Rohit Goyal
- Department of Neuropharmacology, School of Pharmaceutical Sciences, Shoolini University, Himachal Pradesh, India.
| |
Collapse
|
10
|
Araki T, Hiragi T, Kuga N, Luo C, Andoh M, Sugao K, Nagata H, Sasaki T, Ikegaya Y, Koyama R. Microglia induce auditory dysfunction after status epilepticus in mice. Glia 2024; 72:274-288. [PMID: 37746760 DOI: 10.1002/glia.24472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 09/26/2023]
Abstract
Auditory dysfunction and increased neuronal activity in the auditory pathways have been reported in patients with temporal lobe epilepsy, but the cellular mechanisms involved are unknown. Here, we report that microglia play a role in the disinhibition of auditory pathways after status epilepticus in mice. We found that neuronal activity in the auditory pathways, including the primary auditory cortex and the medial geniculate body (MGB), was increased and auditory discrimination was impaired after status epilepticus. We further demonstrated that microglia reduced inhibitory synapses on MGB relay neurons over an 8-week period after status epilepticus, resulting in auditory pathway hyperactivity. In addition, we found that local removal of microglia from the MGB attenuated the increase in c-Fos+ relay neurons and improved auditory discrimination. These findings reveal that thalamic microglia are involved in auditory dysfunction in epilepsy.
Collapse
Affiliation(s)
- Tasuku Araki
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Toshimitsu Hiragi
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Nahoko Kuga
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Cong Luo
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Megumi Andoh
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | | | | | - Takuya Sasaki
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- Institute for AI and Beyond, The University of Tokyo, Tokyo, Japan
- Center for Information and Neural Networks, National Institute of Information and Communications Technology, Osaka, Japan
| | - Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- Institute for AI and Beyond, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
O’Day DH. The Complex Interplay between Toxic Hallmark Proteins, Calmodulin-Binding Proteins, Ion Channels, and Receptors Involved in Calcium Dyshomeostasis in Neurodegeneration. Biomolecules 2024; 14:173. [PMID: 38397410 PMCID: PMC10886625 DOI: 10.3390/biom14020173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 01/23/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Calcium dyshomeostasis is an early critical event in neurodegeneration as exemplified by Alzheimer's (AD), Huntington's (HD) and Parkinson's (PD) diseases. Neuronal calcium homeostasis is maintained by a diversity of ion channels, buffers, calcium-binding protein effectors, and intracellular storage in the endoplasmic reticulum, mitochondria, and lysosomes. The function of these components and compartments is impacted by the toxic hallmark proteins of AD (amyloid beta and Tau), HD (huntingtin) and PD (alpha-synuclein) as well as by interactions with downstream calcium-binding proteins, especially calmodulin. Each of the toxic hallmark proteins (amyloid beta, Tau, huntingtin, and alpha-synuclein) binds to calmodulin. Multiple channels and receptors involved in calcium homeostasis and dysregulation also bind to and are regulated by calmodulin. The primary goal of this review is to show the complexity of these interactions and how they can impact research and the search for therapies. A secondary goal is to suggest that therapeutic targets downstream from calcium dyshomeostasis may offer greater opportunities for success.
Collapse
Affiliation(s)
- Danton H. O’Day
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada;
- Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| |
Collapse
|
12
|
Bosetti C, Ferrini L, Ferrari AR, Bartolini E, Calderoni S. Children with Autism Spectrum Disorder and Abnormalities of Clinical EEG: A Qualitative Review. J Clin Med 2024; 13:279. [PMID: 38202286 PMCID: PMC10779511 DOI: 10.3390/jcm13010279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/22/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024] Open
Abstract
Over the last decade, the comorbidity between Autism Spectrum Disorder (ASD) and epilepsy has been widely demonstrated, and many hypotheses regarding the common neurobiological bases of these disorders have been put forward. A variable, but significant, prevalence of abnormalities on electroencephalogram (EEG) has been documented in non-epileptic children with ASD; therefore, several scientific studies have recently tried to demonstrate the role of these abnormalities as a possible biomarker of altered neural connectivity in ASD individuals. This narrative review intends to summarize the main findings of the recent scientific literature regarding abnormalities detected with standard EEG in children/adolescents with idiopathic ASD. Research using three different databases (PubMed, Scopus and Google Scholar) was conducted, resulting in the selection of 10 original articles. Despite an important lack of studies on preschoolers and a deep heterogeneity in results, some authors speculated on a possible association between EEG abnormalities and ASD characteristics, in particular, the severity of symptoms. Although this correlation needs to be more strongly elucidated, these findings may encourage future studies aimed at demonstrating the role of electrical brain abnormalities as an early biomarker of neural circuit alterations in ASD, highlighting the potential diagnostic, prognostic and therapeutic value of EEG in this field.
Collapse
Affiliation(s)
- Chiara Bosetti
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (C.B.); (L.F.); (A.R.F.); (S.C.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Luca Ferrini
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (C.B.); (L.F.); (A.R.F.); (S.C.)
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
| | - Anna Rita Ferrari
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (C.B.); (L.F.); (A.R.F.); (S.C.)
| | - Emanuele Bartolini
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (C.B.); (L.F.); (A.R.F.); (S.C.)
- Tuscany PhD Programme in Neurosciences, 50139 Florence, Italy
| | - Sara Calderoni
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (C.B.); (L.F.); (A.R.F.); (S.C.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
13
|
Subramaniam S, Boregowda S. Curbing Rhes Actions: Mechanism-based Molecular Target for Huntington's Disease and Tauopathies. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:21-29. [PMID: 36959146 DOI: 10.2174/1871527322666230320103518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 03/25/2023]
Abstract
A highly interconnected network of diverse brain regions is necessary for the precise execution of human behaviors, including cognitive, psychiatric, and motor functions. Unfortunately, degeneration of specific brain regions causes several neurodegenerative disorders, but the mechanisms that elicit selective neuronal vulnerability remain unclear. This knowledge gap greatly hinders the development of effective mechanism-based therapies, despite the desperate need for new treatments. Here, we emphasize the importance of the Rhes (Ras homolog-enriched in the striatum) protein as an emerging therapeutic target. Rhes, an atypical small GTPase with a SUMO (small ubiquitin-like modifier) E3-ligase activity, modulates biological processes such as dopaminergic transmission, alters gene expression, and acts as an inhibitor of motor stimuli in the brain striatum. Mutations in the Rhes gene have also been identified in selected patients with autism and schizophrenia. Moreover, Rhes SUMOylates pathogenic form of mutant huntingtin (mHTT) and tau, enhancing their solubility and cell toxicity in Huntington's disease and tauopathy models. Notably, Rhes uses membrane projections resembling tunneling nanotubes to transport mHTT between cells and Rhes deletion diminishes mHTT spread in the brain. Thus, we predict that effective strategies aimed at diminishing brain Rhes levels will prevent or minimize the abnormalities that occur in HD and tauopathies and potentially in other brain disorders. We review the emerging technologies that enable specific targeting of Rhes in the brain to develop effective disease-modifying therapeutics.
Collapse
Affiliation(s)
- Srinivasa Subramaniam
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, C323, Florida, Jupiter, 33458, USA
| | - Siddaraju Boregowda
- Department of Molecular Therapeutics, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, C323, Florida, Jupiter, 33458, USA
| |
Collapse
|
14
|
Bonnin EA, Golmohammadi A, Rehm R, Tetzlaff C, Rizzoli SO. High-resolution analysis of bound Ca 2+ in neurons and synapses. Life Sci Alliance 2024; 7:e202302030. [PMID: 37833073 PMCID: PMC10575792 DOI: 10.26508/lsa.202302030] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
Calcium (Ca2+) is a well-known second messenger in all cells, and is especially relevant for neuronal activity. Neuronal Ca2+ is found in different forms, with a minority being freely soluble in the cell and more than 99% being bound to proteins. Free Ca2+ has received much attention over the last few decades, but protein-bound Ca2+ has been difficult to analyze. Here, we introduce correlative fluorescence and nanoscale secondary ion mass spectrometry imaging as a tool to describe bound Ca2+ As expected, bound Ca2+ is ubiquitous. It does not correlate to free Ca2+ dynamics at the whole-neuron level, but does correlate significantly to the intensity of markers for GABAergic pre-synapse and glutamatergic post-synapses. In contrast, a negative correlation to pre-synaptic activity was observed, with lower levels of bound Ca2+ observed in the more active synapses. We conclude that bound Ca2+ may regulate neuronal activity and should receive more attention in the future.
Collapse
Affiliation(s)
- Elisa A Bonnin
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
- Excellence Cluster Multiscale Bioimaging (MBExC), Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, Göttingen, Germany
| | - Arash Golmohammadi
- Group of Computational Synaptic Physiology, Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Ronja Rehm
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Christian Tetzlaff
- Group of Computational Synaptic Physiology, Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Silvio O Rizzoli
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
- Excellence Cluster Multiscale Bioimaging (MBExC), Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
15
|
Veshchitskii A, Merkulyeva N. Calcium-binding protein parvalbumin in the spinal cord and dorsal root ganglia. Neurochem Int 2023; 171:105634. [PMID: 37967669 DOI: 10.1016/j.neuint.2023.105634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 09/20/2023] [Accepted: 10/26/2023] [Indexed: 11/17/2023]
Abstract
Parvalbumin is one of the calcium-binding proteins. In the spinal cord, it is mainly expressed in inhibitory neurons; in the dorsal root ganglia, it is expressed in proprioceptive neurons. In contrast to in the brain, weak systematization of parvalbumin-expressing neurons occurs in the spinal cord. The aim of this paper is to provide a systematic review of parvalbumin-expressing neuronal populations throughout the spinal cord and the dorsal root ganglia of mammals, regarding their mapping, co-expression with some functional markers. The data reviewed are mostly concerning rodentia species because they are predominantly presented in literature.
Collapse
Affiliation(s)
- Aleksandr Veshchitskii
- Neuromorphology Lab, Pavlov Institute of Physiology Russian Academy of Sciences, Saint Petersburg, Russia
| | - Natalia Merkulyeva
- Neuromorphology Lab, Pavlov Institute of Physiology Russian Academy of Sciences, Saint Petersburg, Russia.
| |
Collapse
|
16
|
Wang J, Tang M, Xie X, Xu Y, Su P, Jin Z. Efficacy of ferulic acid in the treatment of acute ischemic stroke injury in rats: a systematic review and meta-analysis. Front Pharmacol 2023; 14:1278036. [PMID: 37927604 PMCID: PMC10620722 DOI: 10.3389/fphar.2023.1278036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/10/2023] [Indexed: 11/07/2023] Open
Abstract
Background: Intravenous thrombolysis is commonly used in the treatment of acute ischemic stroke damage. The existing thrombolytic drugs still suffer significant shortcomings, including a limited fibrin specificity and bleeding complications. Ferulic acid can directly bind the key thrombus enzymes and target to blood clots, suggesting its thrombolytic potency that may be beneficial with thrombolytic potency for the treatment of acute ischemic stroke damage. Objective: The purpose of this systematic review and meta-analysis was to evaluate the efficacy of ferulic acid in the treatment of acute ischemic stroke injury in rats and its potential mechanism of action. Materials and methods: We conducted a literature search in six databases, including CNKI, up to July 2023. Results: Sixteen trials were included in the meta-analysis, which demonstrated that ferulic acid significantly reduced infarct size, neurological deficit score, apoptosis index, cleaved caspase-3, and cytochrome C levels (all p < 0.05). In addition, ferulic acid significantly increased the levels of phosphorylated Akt, mitochondrial Bcl-xL/Bax, phosphorylated astrocyte PEA15, hippocampal calcium binding protein, and mitochondrial Bcl-2/Bax ratio (all p < 0.05). Conclusion: This study demonstrates that ferulic acid protects against acute ischemic stroke injury in rats by inhibiting ischemia-induced excitotoxicity, inflammatory response, and apoptosis.
Collapse
Affiliation(s)
- Jiashan Wang
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Meiqi Tang
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Xiuzhen Xie
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yingqi Xu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Pingping Su
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhuqing Jin
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
17
|
Aragona M, Briglia M, Porcino C, Mhalhel K, Cometa M, Germanà PG, Montalbano G, Levanti M, Laurà R, Abbate F, Germanà A, Guerrera MC. Localization of Calretinin, Parvalbumin, and S100 Protein in Nothobranchius guentheri Retina: A Suitable Model for the Retina Aging. Life (Basel) 2023; 13:2050. [PMID: 37895432 PMCID: PMC10608213 DOI: 10.3390/life13102050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/05/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Calcium-binding proteins (CaBPs) are members of a heterogeneous family of proteins able to buffer intracellular Ca2+ ion concentration. CaBPs are expressed in the central and peripheral nervous system, including a subpopulation of retinal neurons. Since neurons expressing different CaBPs show different susceptibility to degeneration, it could be hypothesized that they are not just markers of different neuronal subpopulations, but that they might be crucial in survival. CaBPs' ability to buffer Ca2+ cytoplasmatic concentration makes them able to defend against a toxic increase in intracellular calcium that can lead to neurodegenerative processes, including those related to aging. An emergent model for aging studies is the annual killifish belonging to the Nothobranchius genus, thanks to its short lifespan. Members of this genus, such as Nothobranchius guentheri, show a retinal stratigraphy similar to that of other actinopterygian fishes and humans. However, according to our knowledge, CaBPs' occurrence and distribution in the retina of N. guentheri have never been investigated before. Therefore, the present study aimed to localize Calretinin N-18, Parvalbumin, and S100 protein (S100p) in the N. guentheri retina with immunohistochemistry methods. The results of the present investigation demonstrate for the first time the occurrence of Calretinin N-18, Parvalbumin, and S100p in N. guentheri retina and, consequently, the potential key role of these CaBPs in the biology of the retinal cells. Hence, the suitability of N. guentheri as a model to study the changes in CaBPs' expression patterns during neurodegenerative processes affecting the retina related both to disease and aging can be assumed.
Collapse
Affiliation(s)
| | | | - Caterina Porcino
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (M.A.); (M.B.); (K.M.); (M.C.); (P.G.G.); (G.M.); (M.L.); (R.L.); (F.A.); (A.G.); (M.C.G.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Porseva VV, Preobrazhensky ND. Neuronal nitric oxide synthase and calbindin expression in sympathetic preganglionic neurons following capsaicin treatment. Anat Rec (Hoboken) 2023; 306:2264-2275. [PMID: 35717590 DOI: 10.1002/ar.25012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/19/2022] [Accepted: 05/13/2022] [Indexed: 11/08/2022]
Abstract
Along with well-known data on the neurochemical mechanisms of nociceptor activation, there are still no clear data regarding changes in the cellular composition and morphological characteristics of spinal preganglionic neurons (SPN) after capsaicin treatment. The mechanism of capsaicin toxicity differs in developing and mature nerve cells. This study aimed to determine the number of SPN in the autonomic nuclei on spinal cord (SC) sections and their cross-sectional area, the localization, percentage, and profile area of SPN containing neuronal nitric oxide synthase (nNOS) and calbindin (CB) in the thoracic SC of rats of different ages (from birth to 1-year-old) after capsaicin treatment. Neonatal capsaicin treatment generally decreased the cross-sectional area of the SPN pericarya. However, the cross-sectional area of the CB-immunoreactive (IR) SPN increased in the central autonomic area in rats aged 10-30 days old after capsaicin treatment. The number of SPN decreased only in the central autonomic area of rats aged <20 days. The proportion of nNOS-IR neurons remained steady and did not change during development. The cross-sectional area of nNOS-IR SPN in capsaicin-treated rats was less than that in control rats. The results obtained will promote further studies on the mechanisms of sensory processing in the SC and the development of the sympathetic nervous system.
Collapse
Affiliation(s)
- Valentina V Porseva
- Department of Pathophysiology, Yaroslavl State Medical University, Yaroslavl, Russia
| | | |
Collapse
|
19
|
Téllez de Meneses PG, Pérez-Revuelta L, Canal-Alonso Á, Hernández-Pérez C, Cocho T, Valero J, Weruaga E, Díaz D, Alonso JR. Immunohistochemical distribution of secretagogin in the mouse brain. Front Neuroanat 2023; 17:1224342. [PMID: 37711587 PMCID: PMC10498459 DOI: 10.3389/fnana.2023.1224342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/09/2023] [Indexed: 09/16/2023] Open
Abstract
Introduction Calcium is essential for the correct functioning of the central nervous system, and calcium-binding proteins help to finely regulate its concentration. Whereas some calcium-binding proteins such as calmodulin are ubiquitous and are present in many cell types, others such as calbindin, calretinin, and parvalbumin are expressed in specific neuronal populations. Secretagogin belongs to this latter group and its distribution throughout the brain is only partially known. In the present work, the distribution of secretagogin-immunopositive cells was studied in the entire brain of healthy adult mice. Methods Adult male C57BL/DBA mice aged between 5 and 7 months were used. Their whole brain was sectioned and used for immunohistochemistry. Specific neural populations were observed in different zones and nuclei identified according to Paxinos mouse brain atlas. Results Labelled cells were found with a Golgi-like staining, allowing an excellent characterization of their dendritic and axonal arborizations. Many secretagogin-positive cells were observed along different encephalic regions, especially in the olfactory bulb, basal ganglia, and hypothalamus. Immunostained populations were very heterogenous in both size and distribution, as some nuclei presented labelling in their entire extension, but in others, only scattered cells were present. Discussion Secretagogin can provide a more complete vision of calcium-buffering mechanisms in the brain, and can be a useful neuronal marker in different brain areas for specific populations.
Collapse
Affiliation(s)
- Pablo G. Téllez de Meneses
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Laura Pérez-Revuelta
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Ángel Canal-Alonso
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Bioinformatics, Intelligent Systems and Educational Technology (BISITE) Research Group, Universidad de Salamanca, Salamanca, Spain
| | - Carlos Hernández-Pérez
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Teresa Cocho
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Jorge Valero
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Eduardo Weruaga
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - David Díaz
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - José R. Alonso
- Institute for Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| |
Collapse
|
20
|
Rengifo AC, Rivera J, Álvarez-Díaz DA, Naizaque J, Santamaria G, Corchuelo S, Gómez CY, Torres-Fernández O. Morphological and Molecular Changes in the Cortex and Cerebellum of Immunocompetent Mice Infected with Zika Virus. Viruses 2023; 15:1632. [PMID: 37631975 PMCID: PMC10458311 DOI: 10.3390/v15081632] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Zika virus (ZIKV) disease continues to be a threat to public health, and it is estimated that millions of people have been infected and that there have been more cases of serious complications than those already reported. Despite many studies on the pathogenesis of ZIKV, several of the genes involved in the malformations associated with viral infection are still unknown. In this work, the morphological and molecular changes in the cortex and cerebellum of mice infected with ZIKV were evaluated. Neonatal BALB/c mice were inoculated with ZIKV intraperitoneally, and the respective controls were inoculated with a solution devoid of the virus. At day 10 postinoculation, the mice were euthanized to measure the expression of the markers involved in cortical and cerebellar neurodevelopment. The infected mice presented morphological changes accompanied by calcifications, as well as a decrease in most of the markers evaluated in the cortex and cerebellum. The modifications found could be predictive of astrocytosis, dendritic pathology, alterations in the regulation systems of neuronal excitation and inhibition, and premature maturation, conditions previously described in other models of ZIKV infection and microcephaly.
Collapse
Affiliation(s)
- Aura Caterine Rengifo
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Jorge Rivera
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Diego Alejandro Álvarez-Díaz
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
- Genómica de Microorganismos Emergentes, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia
| | - Julián Naizaque
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Gerardo Santamaria
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Sheryll Corchuelo
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Claudia Yadira Gómez
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Orlando Torres-Fernández
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| |
Collapse
|
21
|
Wong KC, Jayapalan JJ, Subramanian P, Ismail MN, Abdul-Rahman PS. Label-free quantitative mass spectrometry analysis of the circadian proteome of Drosophila melanogaster lethal giant larvae mutants reveals potential therapeutic effects of melatonin. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2023; 113:e22008. [PMID: 36915983 DOI: 10.1002/arch.22008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/10/2023] [Accepted: 02/22/2023] [Indexed: 05/16/2023]
Abstract
Mutation in the Drosophila melanogaster lethal giant larvae (lgl), a tumor suppressor gene with a well-established role in cellular polarity, is known to results in massive cellular proliferation and neoplastic outgrowths. Although the tumorigenic properties of lgl mutant have been previously studied, however, little is known about its consequences on the proteome. In this study, mass spectrometry-based label-free quantitative proteomics was employed to investigate the changes in the head and intestinal tissues proteins of Drosophila melanogaster, due to lgl mutation and following treatment with melatonin. Additionally, to uncover the time-influenced variations in the proteome during tumorigenesis and melatonin treatment, the rhythmic expression of proteins was also investigated at 6-h intervals within 24-h clock. Together, the present study has identified 434 proteins of altered expressions (p < 0.05 and fold change ±1.5) in the tissues of flies in response to lgl mutation as well as posttreatment with melatonin. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of differentially expressed proteins revealed that lgl mutation had significantly affected the biological functions, including metabolism, and protein synthesis and degradation, in flies' tissues. Besides, melatonin had beneficially mitigated the deleterious effects of lgl mutation by reversing the alterations in protein expression closer to baseline levels. Further, changes in protein expression in the tissues due to lgl mutation and melatonin treatment were found rhythmically orchestrated. Together, these findings provide novel insight into the pathways involved in lgl-induced tumorigenesis as well as demonstrated the efficacy of melatonin as a potential anticancer agent. Data are available via ProteomeXchange with identifier PXD033191.
Collapse
Affiliation(s)
- Kar-Cheng Wong
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Jaime J Jayapalan
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- Universiti Malaya Centre for Proteomics Research (UMCPR), Universiti Malaya, Kuala Lumpur, Malaysia
| | - Perumal Subramanian
- Department of Biochemistry and Biotechnology, Annamalai University, Chidambaram, Tamil Nadu, India
| | - Mohd Nazri Ismail
- Analytical Biochemistry Research Centre, Universiti Sains Malaysia, Bayan Lepas, Penang, Malaysia
| | - Puteri S Abdul-Rahman
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- Universiti Malaya Centre for Proteomics Research (UMCPR), Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
22
|
Yang X, Tohda C. Diosgenin restores memory function via SPARC-driven axonal growth from the hippocampus to the PFC in Alzheimer's disease model mice. Mol Psychiatry 2023; 28:2398-2411. [PMID: 37085711 PMCID: PMC10611574 DOI: 10.1038/s41380-023-02052-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 04/23/2023]
Abstract
Central nervous system axons have minimal capacity to regenerate in adult brains, hindering memory recovery in Alzheimer's disease (AD). Although recent studies have shown that damaged axons sprouted in adult and AD mouse brains, long-distance axonal re-innervation to their targets has not been achieved. We selectively visualized axon-growing neurons in the neural circuit for memory formation, from the hippocampus to the prefrontal cortex, and showed that damaged axons successfully extended to their native projecting area in mouse models of AD (5XFAD) by administration of an axonal regenerative agent, diosgenin. In vivo transcriptome analysis detected the expression profile of axon-growing neurons directly isolated from the hippocampus of 5XFAD mice. Secreted protein acidic and rich in cysteine (SPARC) was the most expressed gene in axon-growing neurons. Neuron-specific overexpression of SPARC via adeno-associated virus serotype 9 delivery in the hippocampus recovered memory deficits and axonal projection to the prefrontal cortex in 5XFAD mice. DREADDs (Designer receptors exclusively activated by designer drugs) analyses revealed that SPARC overexpression-induced axonal growth in the 5XFAD mouse brain directly contributes to memory recovery. Elevated levels of SPARC on axonal membranes interact with extracellular rail-like collagen type I to promote axonal remodeling along their original tracings in primary cultured hippocampal neurons. These findings suggest that SPARC-driven axonal growth in the brain may be a promising therapeutic strategy for AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Ximeng Yang
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Chihiro Tohda
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| |
Collapse
|
23
|
Retinoic Acid Prevents the Neuronal Damage Through the Regulation of Parvalbumin in an Ischemic Stroke Model. Neurochem Res 2023; 48:487-501. [PMID: 36245066 DOI: 10.1007/s11064-022-03769-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/22/2022] [Accepted: 09/24/2022] [Indexed: 02/04/2023]
Abstract
Ischemic stroke is a neurological disease that causes brain damage by increasing oxidative stress and ion imbalance. Retinoic acid is a major metabolite of vitamin A and regulates oxidative stress, calcium homeostasis, and cell death. Intracellular calcium is involved in neuronal growth and synaptic plasticity. Parvalbumin is a calcium-binding protein that is mainly expressed in brain. In this study, we investigated whether retinoic acid has neuroprotective effects by controlling intracellular calcium concentration and parvalbumin expression in ischemic brain damage. Middle cerebral artery occlusion (MCAO) was performed to induce cerebral ischemia. Retinoic acid (5 mg/kg) or vehicle was injected into the abdominal cavity for four days before surgery and cerebral cortices were collected 24 h after MCAO for further studies. MCAO damage induced neurological deficits and histopathological changes and decreased parvalbumin expression. However, retinoic acid treatment alleviated these changes. In cultured neurons, glutamate (5 mM) exposure induced neuronal cell death, increased intracellular calcium concentration, and decreased parvalbumin expression. Retinoic acid treatment attenuated these changes against glutamate toxicity in a dose-dependent manner. It also regulates glutamate induced change in bcl-2 and bax expression. The mitigation effects of retinoic acid were greater under non-transfection conditions than under parvalbumin siRNA transfection conditions. Our findings showed that retinoic acid modulates intracellular calcium concentration and parvalbumin expression and prevents apoptosis in ischemic brain injury. In conclusion, retinoic acid contributes to the preservation of neurons from ischemic stroke by controlling parvalbumin expression and apoptosis-related proteins.
Collapse
|
24
|
Huang Z, Sun L, Zheng X, Zhang Y, Zhu Y, Chen T, Chen Z, Ja L, OuYang L, Zhu Y, Chen S, Lei W. A neural tract tracing study on synaptic connections for cortical glutamatergic terminals and cervical spinal calretinin neurons in rats. Front Neural Circuits 2023; 17:1086873. [PMID: 37187913 PMCID: PMC10175624 DOI: 10.3389/fncir.2023.1086873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
The cerebral cortex innervates motor neurons in the anterior horn of the spinal cord by regulating of interneurons. At present, nerve tracing, immunohistochemistry, and immunoelectron microscopy are used to explore and confirm the characteristics of synaptic connections between the corticospinal tract (CST) and cervical spinal calretinin (Cr) interneurons. Our morphological results revealed that (1) biotinylated dextran amine labeled (BDA+) fibers from the cerebral cortex primarily presented a contralateral spinal distribution, with a denser distribution in the ventral horn (VH) than in the dorsal horn (DH). An electron microscope (EM) showed that BDA+ terminals formed asymmetric synapses with spinal neurons, and their mean labeling rate was not different between the DH and VH. (2) Cr-immunoreactive (Cr+) neurons were unevenly distributed throughout the spinal gray matter, and were denser and larger in the VH than in the DH. At the single labeling electron microscope (EM) level, the labeling rate of Cr+ dendrites was higher in the VH than in the DH, in which Cr+ dendrites mainly received asymmetric synaptic inputs, and between the VH and DH. (3) Immunofluorescence triple labeling showed obvious apposition points among BDA+ terminals, synaptophysin and Cr+ dendrites, with a higher density in the VH than in the DH. (4) Double labeling in EM, BDA+ terminals and Cr+ dendrites presented the same pattern, BDA+ terminals formed asymmetric synapses either with Cr+ dendrites or Cr negative (Cr-) dendrites, and Cr+ dendrites received either BDA+ terminals or BDA- synaptic inputs. The average percentage of BDA+ terminals targeting Cr+ dendrites was higher in the VH than in the DH, but the percentage of BDA+ terminals targeting Cr- dendrites was prominently higher than that targeting Cr+ dendrites. There was no difference in BDA+ terminal size. The percentage rate for Cr+ dendrites receiving BDA+ terminal inputs was lower than that receiving BDA- terminal inputs, and the BDA+ terminal size was larger than the BDA- terminal size received by Cr+ dendrites. The present morphological results suggested that spinal Cr+ interneurons are involved in the regulatory process of the cortico-spinal pathway.
Collapse
Affiliation(s)
- Ziyun Huang
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Liping Sun
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xuefeng Zheng
- Neuroscience Laboratory for Cognitive and Developmental Disorders, Department of Anatomy, Medical College of Jinan University, Guangzhou, China
| | - Ye Zhang
- Neuroscience Laboratory for Cognitive and Developmental Disorders, Department of Anatomy, Medical College of Jinan University, Guangzhou, China
| | - Yaxi Zhu
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tao Chen
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhi Chen
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Linju Ja
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lisi OuYang
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yaofeng Zhu
- College of Medicine, Institute of Medical Sciences, Jishou University, Jishou, China
- Yaofeng Zhu, ,
| | - Si Chen
- Department of Human Anatomy, Histology and Embryology, Zunyi Medical University, Zhuhai, China
- Si Chen, ,
| | - Wanlong Lei
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Wanlong Lei, ,
| |
Collapse
|
25
|
La Barbera L, Nobili A, Cauzzi E, Paoletti I, Federici M, Saba L, Giacomet C, Marino R, Krashia P, Melone M, Keller F, Mercuri NB, Viscomi MT, Conti F, D’Amelio M. Upregulation of Ca 2+-binding proteins contributes to VTA dopamine neuron survival in the early phases of Alzheimer's disease in Tg2576 mice. Mol Neurodegener 2022; 17:76. [PMID: 36434727 PMCID: PMC9700939 DOI: 10.1186/s13024-022-00580-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 10/31/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Recent clinical and experimental studies have highlighted the involvement of Ventral Tegmental Area (VTA) dopamine (DA) neurons for the early pathogenesis of Alzheimer's Disease (AD). We have previously described a progressive and selective degeneration of these neurons in the Tg2576 mouse model of AD, long before amyloid-beta plaque formation. The degenerative process in DA neurons is associated with an autophagy flux impairment, whose rescue can prevent neuronal loss. Impairments in autophagy can be the basis for accumulation of damaged mitochondria, leading to disturbance in calcium (Ca2+) homeostasis, and to functional and structural deterioration of DA neurons. METHODS In Tg2576 mice, we performed amperometric recordings of DA levels and analysis of dopaminergic fibers in the Nucleus Accumbens - a major component of the ventral striatum precociously affected in AD patients - together with retrograde tracing, to identify the most vulnerable DA neuron subpopulations in the VTA. Then, we focused on these neurons to analyze mitochondrial integrity and Apoptosis-inducing factor (AIF) localization by electron and confocal microscopy, respectively. Stereological cell count was also used to evaluate degeneration of DA neuron subpopulations containing the Ca2+-binding proteins Calbindin-D28K and Calretinin. The expression levels for these proteins were analyzed by western blot and confocal microscopy. Lastly, using electrophysiology and microfluorometry we analyzed VTA DA neuron intrinsic properties and cytosolic free Ca2+ levels. RESULTS We found a progressive degeneration of mesolimbic DA neurons projecting to the ventral striatum, located in the paranigral nucleus and parabrachial pigmented subnucleus of the VTA. At the onset of degeneration (3 months of age), the vulnerable DA neurons in the Tg2576 accumulate damaged mitochondria, while AIF translocates from the mitochondria to the nucleus. Although we describe an age-dependent loss of the DA neurons expressing Calbindin-D28K or Calretinin, we observed that the remaining cells upregulate the levels of Ca2+-binding proteins, and the free cytosolic levels of Ca2+ in these neurons are significantly decreased. Coherently, TUNEL-stained Tg2576 DA neurons express lower levels of Calbindin-D28K when compared with non-apoptotic cells. CONCLUSION Overall, our results suggest that the overexpression of Ca2+-binding proteins in VTA DA neurons might be an attempt of cells to survive by increasing their ability to buffer free Ca2+. Exploring strategies to overexpress Ca2+-binding proteins could be fundamental to reduce neuronal suffering and improve cognitive and non-cognitive functions in AD.
Collapse
Affiliation(s)
- Livia La Barbera
- grid.9657.d0000 0004 1757 5329Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy ,grid.417778.a0000 0001 0692 3437Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Annalisa Nobili
- grid.9657.d0000 0004 1757 5329Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy ,grid.417778.a0000 0001 0692 3437Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Emma Cauzzi
- grid.417778.a0000 0001 0692 3437Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143 Rome, Italy ,grid.6530.00000 0001 2300 0941Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Ilaria Paoletti
- grid.9657.d0000 0004 1757 5329Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Mauro Federici
- grid.417778.a0000 0001 0692 3437Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Luana Saba
- grid.9657.d0000 0004 1757 5329Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy ,grid.417778.a0000 0001 0692 3437Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Cecilia Giacomet
- grid.6530.00000 0001 2300 0941Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Ramona Marino
- grid.9657.d0000 0004 1757 5329Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Paraskevi Krashia
- grid.417778.a0000 0001 0692 3437Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143 Rome, Italy ,grid.9657.d0000 0004 1757 5329Department of Sciences and Technologies for Humans and Environment, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Marcello Melone
- grid.7010.60000 0001 1017 3210Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche (UNIVPM), 60020 Ancona, Italy ,Center for Neurobiology of Aging, IRCCS Istituto Nazionale Ricovero e Cura Anziani (INRCA), 60020 Ancona, Italy
| | - Flavio Keller
- grid.9657.d0000 0004 1757 5329Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Nicola Biagio Mercuri
- grid.417778.a0000 0001 0692 3437Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143 Rome, Italy ,grid.6530.00000 0001 2300 0941Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Maria Teresa Viscomi
- grid.8142.f0000 0001 0941 3192Department of Life Science and Public Health; Section of Histology and Embryology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy ,grid.414603.4Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy
| | - Fiorenzo Conti
- grid.7010.60000 0001 1017 3210Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche (UNIVPM), 60020 Ancona, Italy ,Center for Neurobiology of Aging, IRCCS Istituto Nazionale Ricovero e Cura Anziani (INRCA), 60020 Ancona, Italy ,grid.7010.60000 0001 1017 3210Foundation for Molecular Medicine, Università Politecnica delle Marche, 60020 Ancona, Italy
| | - Marcello D’Amelio
- grid.9657.d0000 0004 1757 5329Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy ,grid.417778.a0000 0001 0692 3437Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| |
Collapse
|
26
|
Quach TT, Stratton HJ, Khanna R, Mackey-Alfonso S, Deems N, Honnorat J, Meyer K, Duchemin AM. Neurodegenerative Diseases: From Dysproteostasis, Altered Calcium Signalosome to Selective Neuronal Vulnerability to AAV-Mediated Gene Therapy. Int J Mol Sci 2022; 23:ijms232214188. [PMID: 36430666 PMCID: PMC9694178 DOI: 10.3390/ijms232214188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022] Open
Abstract
Despite intense research into the multifaceted etiology of neurodegenerative diseases (ND), they remain incurable. Here we provide a brief overview of several major ND and explore novel therapeutic approaches. Although the cause (s) of ND are not fully understood, the accumulation of misfolded/aggregated proteins in the brain is a common pathological feature. This aggregation may initiate disruption of Ca++ signaling, which is an early pathological event leading to altered dendritic structure, neuronal dysfunction, and cell death. Presently, ND gene therapies remain unidimensional, elusive, and limited to modifying one pathological feature while ignoring others. Considering the complexity of signaling cascades in ND, we discuss emerging therapeutic concepts and suggest that deciphering the molecular mechanisms involved in dendritic pathology may broaden the phenotypic spectrum of ND treatment. An innovative multiplexed gene transfer strategy that employs silencing and/or over-expressing multiple effectors could preserve vulnerable neurons before they are lost. Such therapeutic approaches may extend brain health span and ameliorate burdensome chronic disease states.
Collapse
Affiliation(s)
- Tam T. Quach
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
- INSERM U1217/CNRS UMR5310, Université de Lyon, Université Claude Bernard Lyon 1, 69677 Lyon, France
| | | | - Rajesh Khanna
- Department of Molecular Pathobiology, New York University, New York, NY 10010, USA
| | - Sabrina Mackey-Alfonso
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Nicolas Deems
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Jérome Honnorat
- INSERM U1217/CNRS UMR5310, Université de Lyon, Université Claude Bernard Lyon 1, 69677 Lyon, France
- French Reference Center on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, 69677 Lyon, France
- SynatAc Team, Institut NeuroMyoGène, 69677 Lyon, France
| | - Kathrin Meyer
- The Research Institute of Nationwide Children Hospital, Columbus, OH 43205, USA
- Department of Pediatric, The Ohio State University, Columbus, OH 43210, USA
| | - Anne-Marie Duchemin
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: ; Tel.: +1-614-293-5517; Fax: +1-614-293-7599
| |
Collapse
|
27
|
Kwon KM, Pak JH, Jeon CJ. Immunocytochemical localization of the AMPA glutamate receptor subtype GluR2/3 in the squid optic lobe. Acta Histochem 2022; 124:151941. [PMID: 35963117 DOI: 10.1016/j.acthis.2022.151941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/03/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022]
Abstract
As a major excitatory neurotransmitter in the cephalopod visual system, glutamate signaling is facilitated by ionotropic receptors, such as α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors (AMPAR). In cephalopods with large and well-developed brains, the optic lobes (OL) mainly process visual inputs and are involved in learning and memory. Although the presence of AMPAR in squid OL has been reported, the organization of specific AMPAR-containing neurons remains unknown. This study aimed to investigate the immunocytochemical localization of the AMPA glutamate receptor subtype 2/3-immunoreactive (GluR2/3-IR) neurons in the OL of Pacific flying squid (Tordarodes pacificus). Morphologically diverse GluR2/3-IR neurons were predominantly located in the tangential zone of the medulla. Medium-to-large GluR2/3-IR neurons were also detected. The distribution patterns and cell morphologies of calcium-binding protein (CBP)-IR neurons, specifically calbindin-D28K (CB)-, calretinin (CR)-, and parvalbumin (PV)-IR neurons, were similar to those of GluR2/3-IR neurons. However, two-color immunofluorescence revealed that GluR2/3-IR neurons did not colocalize with the CBP-IR neurons. Furthermore, the specific localizations and diverse types of GluR2/3-IR neurons that do not express CB, CR, or PV in squid OL were determined. These findings further contribute to the existing data on glutamatergic visual systems and provide new insights for understanding the visual processing mechanisms in cephalopods.
Collapse
Affiliation(s)
- Kyung-Min Kwon
- Department of Biology, School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, College of Natural Sciences, Brain Science and Engineering Institute, Kyungpook National University, Daegu 41566, Republic of Korea; Research Institute for Dok-do and Ulleung-do Island, Department of Biology, School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jae-Hong Pak
- Research Institute for Dok-do and Ulleung-do Island, Department of Biology, School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Chang-Jin Jeon
- Department of Biology, School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, College of Natural Sciences, Brain Science and Engineering Institute, Kyungpook National University, Daegu 41566, Republic of Korea; Research Institute for Dok-do and Ulleung-do Island, Department of Biology, School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
28
|
Mitra S, Basu S, Singh O, Srivastava A, Singru PS. Calcium-binding proteins typify the dopaminergic neuronal subtypes in the ventral tegmental area of zebra finch, Taeniopygia guttata. J Comp Neurol 2022; 530:2562-2586. [PMID: 35715989 DOI: 10.1002/cne.25352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 11/11/2022]
Abstract
Calcium-binding proteins (CBPs) regulate neuronal function in midbrain dopamine (DA)-ergic neurons in mammals by buffering and sensing the intracellular Ca2+ , and vesicular release. In birds, the equivalent set of neurons are important in song learning, directed singing, courtship, and energy balance, yet the status of CBPs in these neurons is unknown. Herein, for the first time, we probe the nature of CBPs, namely, Calbindin-, Calretinin-, Parvalbumin-, and Secretagogin-expressing DA neurons in the ventral tegmental area (VTA) and substantia nigra (SN) in the midbrain of zebra finch, Taeniopygia guttata. qRT-PCR analysis of ventral midbrain tissue fragment revealed higher Calbindin- and Calretinin-mRNA levels compared to Parvalbumin and Secretagogin. Application of immunofluorescence showed CBP-immunoreactive (-i) neurons in VTA (anterior [VTAa], mid [VTAm], caudal [VTAc]), SN (compacta [SNc], and reticulata [SNr]). Compared to VTAa, higher Calbindin- and Parvalbumin-immunoreactivity (-ir), and lower Calretinin-ir were observed in VTAm and VTAc. Secretagogin-ir was highly localized to VTAa. In SN, Calbindin- and Calretinin-ir were higher in SNc, SNr was Parvalbumin enriched, and Secretagogin-ir was not detected. Weak, moderate, and intense tyrosine hydroxylase (TH)-i VTA neurons were demarcated as subtypes 1, 2, and 3, respectively. While subtype 1 TH-i neurons were neither Calbindin- nor Calretinin-i, ∼80 and ∼65% subtype 2 and ∼30 and ∼45% subtype 3 TH-i neurons co-expressed Calbindin and Calretinin, respectively. All TH-i neuronal subtypes co-expressed Parvalbumin with reciprocal relationship with TH-ir. We suggest that the CBPs may determine VTA DA neuronal heterogeneity and differentially regulate their activity in T. guttata.
Collapse
Affiliation(s)
- Saptarsi Mitra
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Sumela Basu
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Omprakash Singh
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Abhinav Srivastava
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Praful S Singru
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
29
|
Tipton AE, Russek SJ. Regulation of Inhibitory Signaling at the Receptor and Cellular Level; Advances in Our Understanding of GABAergic Neurotransmission and the Mechanisms by Which It Is Disrupted in Epilepsy. Front Synaptic Neurosci 2022; 14:914374. [PMID: 35874848 PMCID: PMC9302637 DOI: 10.3389/fnsyn.2022.914374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
Inhibitory signaling in the brain organizes the neural circuits that orchestrate how living creatures interact with the world around them and how they build representations of objects and ideas. Without tight control at multiple points of cellular engagement, the brain’s inhibitory systems would run down and the ability to extract meaningful information from excitatory events would be lost leaving behind a system vulnerable to seizures and to cognitive decline. In this review, we will cover many of the salient features that have emerged regarding the dynamic regulation of inhibitory signaling seen through the lens of cell biology with an emphasis on the major building blocks, the ligand-gated ion channel receptors that are the first transduction point when the neurotransmitter GABA is released into the synapse. Epilepsy association will be used to indicate importance of key proteins and their pathways to brain function and to introduce novel areas for therapeutic intervention.
Collapse
Affiliation(s)
- Allison E. Tipton
- Graduate Program for Neuroscience, Boston University, Boston, MA, United States
- Biomolecular Pharmacology Program, Boston University School of Medicine, Boston, MA, United States
- Boston University MD/PhD Training Program, Boston, MA, United States
| | - Shelley J. Russek
- Biomolecular Pharmacology Program, Boston University School of Medicine, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
- Center for Systems Neuroscience, Boston University, Boston, MA, United States
- Boston University MD/PhD Training Program, Boston, MA, United States
- *Correspondence: Shelley J. Russek,
| |
Collapse
|
30
|
Tuschl K, White RJ, Trivedi C, Valdivia LE, Niklaus S, Bianco IH, Dadswell C, González-Méndez R, Sealy IM, Neuhauss SCF, Houart C, Rihel J, Wilson SW, Busch-Nentwich EM. Loss of slc39a14 causes simultaneous manganese hypersensitivity and deficiency in zebrafish. Dis Model Mech 2022; 15:dmm044594. [PMID: 35514229 PMCID: PMC9227717 DOI: 10.1242/dmm.044594] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 04/25/2022] [Indexed: 12/15/2022] Open
Abstract
Manganese neurotoxicity is a hallmark of hypermanganesemia with dystonia 2, an inherited manganese transporter defect caused by mutations in SLC39A14. To identify novel potential targets of manganese neurotoxicity, we performed transcriptome analysis of slc39a14-/- mutant zebrafish that were exposed to MnCl2. Differentially expressed genes mapped to the central nervous system and eye, and pathway analysis suggested that Ca2+ dyshomeostasis and activation of the unfolded protein response are key features of manganese neurotoxicity. Consistent with this interpretation, MnCl2 exposure led to decreased whole-animal Ca2+ levels, locomotor defects and changes in neuronal activity within the telencephalon and optic tectum. In accordance with reduced tectal activity, slc39a14-/- zebrafish showed changes in visual phototransduction gene expression, absence of visual background adaptation and a diminished optokinetic reflex. Finally, numerous differentially expressed genes in mutant larvae normalised upon MnCl2 treatment indicating that, in addition to neurotoxicity, manganese deficiency is present either subcellularly or in specific cells or tissues. Overall, we assembled a comprehensive set of genes that mediate manganese-systemic responses and found a highly correlated and modulated network associated with Ca2+ dyshomeostasis and cellular stress. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Karin Tuschl
- UCL GOS Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
- Department of Developmental Neurobiology and MRC Centre for Neurodevelopmental Disorders, IoPPN, Kings College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Richard J. White
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London E1 4NS, UK
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Chintan Trivedi
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Leonardo E. Valdivia
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Camino La Pirámide 5750, Huechuraba 8580745, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Camino La Pirámide 5750, Huechuraba 8580745, Chile
| | - Stephanie Niklaus
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Isaac H. Bianco
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Chris Dadswell
- School of Life Sciences, University of Sussex, Brighton BN1 9QJ, UK
| | | | - Ian M. Sealy
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London E1 4NS, UK
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Stephan C. F. Neuhauss
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Corinne Houart
- Department of Developmental Neurobiology and MRC Centre for Neurodevelopmental Disorders, IoPPN, Kings College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Jason Rihel
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Stephen W. Wilson
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Elisabeth M. Busch-Nentwich
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London E1 4NS, UK
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK
| |
Collapse
|
31
|
Boccuni I, Fairless R. Retinal Glutamate Neurotransmission: From Physiology to Pathophysiological Mechanisms of Retinal Ganglion Cell Degeneration. Life (Basel) 2022; 12:638. [PMID: 35629305 PMCID: PMC9147752 DOI: 10.3390/life12050638] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 12/12/2022] Open
Abstract
Glutamate neurotransmission and metabolism are finely modulated by the retinal network, where the efficient processing of visual information is shaped by the differential distribution and composition of glutamate receptors and transporters. However, disturbances in glutamate homeostasis can result in glutamate excitotoxicity, a major initiating factor of common neurodegenerative diseases. Within the retina, glutamate excitotoxicity can impair visual transmission by initiating degeneration of neuronal populations, including retinal ganglion cells (RGCs). The vulnerability of RGCs is observed not just as a result of retinal diseases but has also been ascribed to other common neurodegenerative and peripheral diseases. In this review, we describe the vulnerability of RGCs to glutamate excitotoxicity and the contribution of different glutamate receptors and transporters to this. In particular, we focus on the N-methyl-d-aspartate (NMDA) receptor as the major effector of glutamate-induced mechanisms of neurodegeneration, including impairment of calcium homeostasis, changes in gene expression and signalling, and mitochondrial dysfunction, as well as the role of endoplasmic reticular stress. Due to recent developments in the search for modulators of NMDA receptor signalling, novel neuroprotective strategies may be on the horizon.
Collapse
Affiliation(s)
- Isabella Boccuni
- Institute for Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany;
| | - Richard Fairless
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany;
- Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
32
|
Ma SY, KWAN KM. Size Anomaly and Alteration of GABAergic Enzymes Expressions in Cerebellum of a Valproic acid Mouse Model of Autism. Behav Brain Res 2022; 428:113896. [DOI: 10.1016/j.bbr.2022.113896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/29/2022] [Accepted: 04/13/2022] [Indexed: 11/02/2022]
|
33
|
Akat E, Yenmiş M, Pombal MA, Molist P, Megías M, Arman S, Veselỳ M, Anderson R, Ayaz D. Comparison of Vertebrate Skin Structure at Class Level: A Review. Anat Rec (Hoboken) 2022; 305:3543-3608. [DOI: 10.1002/ar.24908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/14/2022] [Accepted: 02/21/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Esra Akat
- Ege University, Faculty of Science, Biology Department Bornova, İzmir Turkey
| | - Melodi Yenmiş
- Ege University, Faculty of Science, Biology Department Bornova, İzmir Turkey
| | - Manuel A. Pombal
- Universidade de Vigo, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía‐IBIV Vigo, España
| | - Pilar Molist
- Universidade de Vigo, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía‐IBIV Vigo, España
| | - Manuel Megías
- Universidade de Vigo, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía‐IBIV Vigo, España
| | - Sezgi Arman
- Sakarya University, Faculty of Science and Letters, Biology Department Sakarya Turkey
| | - Milan Veselỳ
- Palacky University, Faculty of Science, Department of Zoology Olomouc Czechia
| | - Rodolfo Anderson
- Departamento de Zoologia, Instituto de Biociências Universidade Estadual Paulista São Paulo Brazil
| | - Dinçer Ayaz
- Ege University, Faculty of Science, Biology Department Bornova, İzmir Turkey
| |
Collapse
|
34
|
Godoy LD, Prizon T, Rossignoli MT, Leite JP, Liberato JL. Parvalbumin Role in Epilepsy and Psychiatric Comorbidities: From Mechanism to Intervention. Front Integr Neurosci 2022; 16:765324. [PMID: 35250498 PMCID: PMC8891758 DOI: 10.3389/fnint.2022.765324] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 01/24/2022] [Indexed: 12/22/2022] Open
Abstract
Parvalbumin is a calcium-binding protein present in inhibitory interneurons that play an essential role in regulating many physiological processes, such as intracellular signaling and synaptic transmission. Changes in parvalbumin expression are deeply related to epilepsy, which is considered one of the most disabling neuropathologies. Epilepsy is a complex multi-factor group of disorders characterized by periods of hypersynchronous activity and hyperexcitability within brain networks. In this scenario, inhibitory neurotransmission dysfunction in modulating excitatory transmission related to the loss of subsets of parvalbumin-expressing inhibitory interneuron may have a prominent role in disrupted excitability. Some studies also reported that parvalbumin-positive interneurons altered function might contribute to psychiatric comorbidities associated with epilepsy, such as depression, anxiety, and psychosis. Understanding the epileptogenic process and comorbidities associated with epilepsy have significantly advanced through preclinical and clinical investigation. In this review, evidence from parvalbumin altered function in epilepsy and associated psychiatric comorbidities were explored with a translational perspective. Some advances in potential therapeutic interventions are highlighted, from current antiepileptic and neuroprotective drugs to cutting edge modulation of parvalbumin subpopulations using optogenetics, designer receptors exclusively activated by designer drugs (DREADD) techniques, transcranial magnetic stimulation, genome engineering, and cell grafting. Creating new perspectives on mechanisms and therapeutic strategies is valuable for understanding the pathophysiology of epilepsy and its psychiatric comorbidities and improving efficiency in clinical intervention.
Collapse
Affiliation(s)
- Lívea Dornela Godoy
- Department of Psychology, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Tamiris Prizon
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Matheus Teixeira Rossignoli
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - João Pereira Leite
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- João Pereira Leite,
| | - José Luiz Liberato
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- *Correspondence: José Luiz Liberato,
| |
Collapse
|
35
|
Bokulić E, Medenica T, Knezović V, Štajduhar A, Almahariq F, Baković M, Judaš M, Sedmak G. The Stereological Analysis and Spatial Distribution of Neurons in the Human Subthalamic Nucleus. Front Neuroanat 2022; 15:749390. [PMID: 34970124 PMCID: PMC8712451 DOI: 10.3389/fnana.2021.749390] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 11/17/2021] [Indexed: 11/13/2022] Open
Abstract
The subthalamic nucleus (STN) is a small, ovoid structure, and an important site of deep brain stimulation (DBS) for the treatment of Parkinson’s disease. Although the STN is a clinically important structure, there are many unresolved issues with regard to it. These issues are especially related to the anatomical subdivision, neuronal phenotype, neuronal composition, and spatial distribution. In this study, we have examined the expression pattern of 8 neuronal markers [nNOS, NeuN, parvalbumin (PV), calbindin (CB), calretinin (CR), FOXP2, NKX2.1, and PAX6] in the adult human STN. All of the examined markers, except CB, were present in the STN. To determine the neuronal density, we have performed stereological analysis on Nissl-stained and immunohistochemical slides of positive markers. The stereology data were also used to develop a three-dimensional map of the spatial distribution of neurons within the STN. The nNOS population exhibited the largest neuronal density. The estimated total number of nNOS STN neurons is 281,308 ± 38,967 (± 13.85%). The STN neuronal subpopulations can be divided into two groups: one with a neuronal density of approximately 3,300 neurons/mm3 and the other with a neuronal density of approximately 2,200 neurons/mm3. The largest density of STN neurons was observed along the ventromedial border of the STN and the density gradually decreased toward the dorsolateral border. In this study, we have demonstrated the presence of 7 neuronal markers in the STN, three of which were not previously described in the human STN. The human STN is a collection of diverse, intermixed neuronal subpopulations, and our data, as far as the cytoarchitectonics is concerned, did not support the tripartite STN subdivision.
Collapse
Affiliation(s)
- Ema Bokulić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Centre of Excellence for Basic, Clinical and Translational Neuroscience, Zagreb, Croatia
| | - Tila Medenica
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Centre of Excellence for Basic, Clinical and Translational Neuroscience, Zagreb, Croatia
| | - Vinka Knezović
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Centre of Excellence for Basic, Clinical and Translational Neuroscience, Zagreb, Croatia
| | - Andrija Štajduhar
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Centre of Excellence for Basic, Clinical and Translational Neuroscience, Zagreb, Croatia.,School of Public Health "Andrija Štampar," University of Zagreb School of Medicine, Zagreb, Croatia
| | - Fadi Almahariq
- Centre of Excellence for Basic, Clinical and Translational Neuroscience, Zagreb, Croatia.,Department of Neurosurgery, Clinical Hospital "Dubrava," Zagreb, Croatia
| | - Marija Baković
- Department of Forensic Medicine, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Miloš Judaš
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Centre of Excellence for Basic, Clinical and Translational Neuroscience, Zagreb, Croatia
| | - Goran Sedmak
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Centre of Excellence for Basic, Clinical and Translational Neuroscience, Zagreb, Croatia
| |
Collapse
|
36
|
Setkowicz Z, Gzielo K, Kielbinski M, Janeczko K. Structural changes in the neocortex as correlates of variations in EEG spectra and seizure susceptibility in rat brains with different degrees of dysplasia. J Comp Neurol 2021; 530:1379-1398. [PMID: 34861050 PMCID: PMC9305260 DOI: 10.1002/cne.25282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 10/26/2021] [Accepted: 11/23/2021] [Indexed: 11/13/2022]
Abstract
Disturbances of the early stages of neurogenesis lead to irreversible changes in the structure of the mature brain and its functional impairment, including increased excitability, which may be the basis for drug‐resistant epilepsy. The range of possible clinical symptoms is as wide as the different stages of disturbed neurogenesis may be. In this study, we used a quadruple model of brain dysplasia by comparing structural and functional disorders in animals whose neurogenesis was disturbed with a single dose of 1 Gy of gamma rays at one of the four stages of neurogenesis, that is, on days 13, 15, 17, or 19 of prenatal development. When reached adulthood, the prenatally irradiated rats received EEG teletransmitter implantation. Thereafter, pilocarpine was administered and significant differences in susceptibility to seizure behavioral symptoms were detected depending on the degree of brain dysplasia. Before, during, and after the seizures significant correlations were found between the density of parvalbumin‐immunopositive neurons located in the cerebral cortex and the intensity of behavioral seizure symptoms or increases in the power of particular EEG bands. Neurons expressing calretinin or NPY showed also dysplasia‐related increases without, however, correlations with parameters of seizure intensity. The results point to significant roles of parvalbumin‐expressing interneurons, and also to expression of NPY—an endogenous anticonvulsant and neuroprotectant reducing susceptibility to seizures and supporting neuronal survival.
Collapse
Affiliation(s)
- Zuzanna Setkowicz
- Laboratory of Experimental Neuropathology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Kraków, Poland
| | - Kinga Gzielo
- Laboratory of Experimental Neuropathology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Kraków, Poland
| | - Michal Kielbinski
- Laboratory of Experimental Neuropathology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Kraków, Poland
| | - Krzysztof Janeczko
- Laboratory of Experimental Neuropathology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
37
|
Fairless R, Bading H, Diem R. Pathophysiological Ionotropic Glutamate Signalling in Neuroinflammatory Disease as a Therapeutic Target. Front Neurosci 2021; 15:741280. [PMID: 34744612 PMCID: PMC8567076 DOI: 10.3389/fnins.2021.741280] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/30/2021] [Indexed: 01/15/2023] Open
Abstract
Glutamate signalling is an essential aspect of neuronal communication involving many different glutamate receptors, and underlies the processes of memory, learning and synaptic plasticity. Despite neuroinflammatory diseases covering a range of maladies with very different biological causes and pathophysiologies, a central role for dysfunctional glutamate signalling is becoming apparent. This is not just restricted to the well-described role of glutamate in mediating neurodegeneration, but also includes a myriad of other influences that glutamate can exert on the vasculature, as well as immune cell and glial regulation, reflecting the ability of neurons to communicate with these compartments in order to couple their activity with neuronal requirements. Here, we discuss the role of pathophysiological glutamate signalling in neuroinflammatory disease, using both multiple sclerosis and Alzheimer's disease as examples, and how current steps are being made to harness our growing understanding of these processes in the development of neuroprotective strategies. This review focuses in particular on N-methyl-D-aspartate (NMDA) and 2-amino-3-(3-hydroxy-5-methylisooxazol-4-yl) propionate (AMPA) type ionotropic glutamate receptors, although metabotropic, G-protein-coupled glutamate receptors may also contribute to neuroinflammatory processes. Given the indispensable roles of glutamate-gated ion channels in synaptic communication, means of pharmacologically distinguishing between physiological and pathophysiological actions of glutamate will be discussed that allow deleterious signalling to be inhibited whilst minimising the disturbance of essential neuronal function.
Collapse
Affiliation(s)
- Richard Fairless
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Ricarda Diem
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
38
|
Abstract
Eye movements are indispensable for visual image stabilization during self-generated and passive head and body motion and for visual orientation. Eye muscles and neuronal control elements are evolutionarily conserved, with novel behavioral repertoires emerging during the evolution of frontal eyes and foveae. The precise execution of eye movements with different dynamics is ensured by morphologically diverse yet complementary sets of extraocular muscle fibers and associated motoneurons. Singly and multiply innervated muscle fibers are controlled by motoneuronal subpopulations with largely selective premotor inputs from task-specific ocular motor control centers. The morphological duality of the neuromuscular interface is matched by complementary biochemical and molecular features that collectively assign different physiological properties to the motor entities. In contrast, the functionality represents a continuum where most motor elements contribute to any type of eye movement, although within preferential dynamic ranges, suggesting that signal transmission and muscle contractions occur within bands of frequency-selective pathways.
Collapse
Affiliation(s)
- Anja K E Horn
- Institute of Anatomy and Cell Biology I, Ludwig-Maximilians-University Munich, 80336 Munich, Germany;
| | - Hans Straka
- Department Biology II, Ludwig-Maximilians-University Munich, 82152 Planegg, Germany
| |
Collapse
|
39
|
Mayadali ÜS, Fleuriet J, Mustari M, Straka H, Horn AKE. Transmitter and ion channel profiles of neurons in the primate abducens and trochlear nuclei. Brain Struct Funct 2021; 226:2125-2151. [PMID: 34181058 PMCID: PMC8354957 DOI: 10.1007/s00429-021-02315-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/04/2021] [Indexed: 01/28/2023]
Abstract
Extraocular motoneurons initiate dynamically different eye movements, including saccades, smooth pursuit and vestibulo-ocular reflexes. These motoneurons subdivide into two main types based on the structure of the neuro-muscular interface: motoneurons of singly-innervated (SIF), and motoneurons of multiply-innervated muscle fibers (MIF). SIF motoneurons are thought to provoke strong and brief/fast muscle contractions, whereas MIF motoneurons initiate prolonged, slow contractions. While relevant for adequate functionality, transmitter and ion channel profiles associated with the morpho-physiological differences between these motoneuron types, have not been elucidated so far. This prompted us to investigate the expression of voltage-gated potassium, sodium and calcium ion channels (Kv1.1, Kv3.1b, Nav1.6, Cav3.1-3.3, KCC2), the transmitter profiles of their presynaptic terminals (vGlut1 and 2, GlyT2 and GAD) and transmitter receptors (GluR2/3, NMDAR1, GlyR1α) using immunohistochemical analyses of abducens and trochlear motoneurons and of abducens internuclear neurons (INTs) in macaque monkeys. The main findings were: (1) MIF and SIF motoneurons express unique voltage-gated ion channel profiles, respectively, likely accounting for differences in intrinsic membrane properties. (2) Presynaptic glutamatergic synapses utilize vGlut2, but not vGlut1. (3) Trochlear motoneurons receive GABAergic inputs, abducens neurons receive both GABAergic and glycinergic inputs. (4) Synaptic densities differ between MIF and SIF motoneurons, with MIF motoneurons receiving fewer terminals. (5) Glutamatergic receptor subtypes differ between MIF and SIF motoneurons. While NMDAR1 is intensely expressed in INTs, MIF motoneurons lack this receptor subtype entirely. The obtained cell-type-specific transmitter and conductance profiles illuminate the structural substrates responsible for differential contributions of neurons in the abducens and trochlear nuclei to eye movements.
Collapse
Affiliation(s)
- Ümit Suat Mayadali
- Institute of Anatomy and Cell Biology, Dept. I, Ludwig-Maximilians-University Munich, Pettenkoferstrasse 11, 80336, Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Jérome Fleuriet
- Washington National Primate Research Center, Department of Ophthalmology, University of Washington Seattle, Seattle, WA, USA
- Intensive Care Unit, Raymond Poincaré Hospital, Assistance Publique-Hôpitaux de Paris, Garches, France
| | - Michael Mustari
- Washington National Primate Research Center, Department of Ophthalmology, University of Washington Seattle, Seattle, WA, USA
| | - Hans Straka
- Department of Biology II, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Anja Kerstin Ellen Horn
- Institute of Anatomy and Cell Biology, Dept. I, Ludwig-Maximilians-University Munich, Pettenkoferstrasse 11, 80336, Munich, Germany.
| |
Collapse
|
40
|
Veshchitskii AA, Musienko PE, Merkulyeva NS. Distribution of Calretinin-Immunopositive Neurons in the Cat Lumbar Spinal Cord. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021040074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
41
|
Moreno DG, Utagawa EC, Arva NC, Schafernak KT, Mufson EJ, Perez SE. Postnatal Cytoarchitecture and Neurochemical Hippocampal Dysfunction in Down Syndrome. J Clin Med 2021; 10:jcm10153414. [PMID: 34362198 PMCID: PMC8347520 DOI: 10.3390/jcm10153414] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 12/11/2022] Open
Abstract
Although the prenatal hippocampus displays deficits in cellular proliferation/migration and volume, which are later associated with memory deficits, little is known about the effects of trisomy 21 on postnatal hippocampal cellular development in Down syndrome (DS). We examined postnatal hippocampal neuronal profiles from autopsies of DS and neurotypical (NTD) neonates born at 38-weeks’-gestation up to children 3 years of age using antibodies against non-phosphorylated (SMI-32) and phosphorylated (SMI-34) neurofilament, calbindin D-28k (Calb), calretinin (Calr), parvalbumin (Parv), doublecortin (DCX) and Ki-67, as well as amyloid precursor protein (APP), amyloid beta (Aβ) and phosphorylated tau (p-tau). Although the distribution of SMI-32-immunoreactive (-ir) hippocampal neurons was similar at all ages in both groups, pyramidal cell apical and basal dendrites were intensely stained in NTD cases. A greater reduction in the number of DCX-ir cells was observed in the hippocampal granule cell layer in DS. Although the distribution of Calb-ir neurons was similar between the youngest and oldest NTD and DS cases, Parv-ir was not detected. Conversely, Calr-ir cells and fibers were observed at all ages in DS, while NTD cases displayed mainly Calr-ir fibers. Hippocampal APP/Aβ-ir diffuse-like plaques were seen in DS and NTD. By contrast, no Aβ1–42 or p-tau profiles were observed. These findings suggest that deficits in hippocampal neurogenesis and pyramidal cell maturation and increased Calr immunoreactivity during early postnatal life contribute to cognitive impairment in DS.
Collapse
Affiliation(s)
- David G. Moreno
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ 85013, USA; (D.G.M.); (E.C.U.); (E.J.M.)
| | - Emma C. Utagawa
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ 85013, USA; (D.G.M.); (E.C.U.); (E.J.M.)
| | - Nicoleta C. Arva
- Department of Pathology and Laboratory Medicine, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA;
| | - Kristian T. Schafernak
- Department of Pathology and Laboratory Medicine, Phoenix Children’s Hospital, Phoenix, AZ 85016, USA;
| | - Elliott J. Mufson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ 85013, USA; (D.G.M.); (E.C.U.); (E.J.M.)
| | - Sylvia E. Perez
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ 85013, USA; (D.G.M.); (E.C.U.); (E.J.M.)
- Correspondence: ; Tel.: +6-02-406-3342
| |
Collapse
|
42
|
Reichenberger I, Caussidier-Dechesne CJ, Straka H. Calretinin Immunoreactivity in the VIIIth Nerve and Inner Ear Endorgans of Ranid Frogs. Front Neurosci 2021; 15:691962. [PMID: 34305520 PMCID: PMC8292642 DOI: 10.3389/fnins.2021.691962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/16/2021] [Indexed: 11/13/2022] Open
Abstract
Calcium-binding proteins are essential for buffering intracellular calcium concentrations, which are critical for regulating cellular processes involved in neuronal computations. One such calcium-binding protein, calretinin, is present in many neurons of the central nervous system as well as those which innervate cranial sensory organs, although often with differential distributions in adjacent cellular elements. Here, we determined the presence and distribution of calretinin-immunoreactivity in the peripheral vestibular and auditory system of ranid frogs. Calretinin-immunoreactivity was observed in ganglion cells innervating the basilar and amphibian papilla, and in a subpopulation of ganglion cells innervating the saccular epithelium. In contrast, none of the ganglion cells innervating the lagena, the utricle, or the three semicircular canals were calretinin-immunopositive, suggesting that this calcium-binding protein is a marker for auditory but not vestibular afferent fibers in the frog. The absence of calretinin in vestibular ganglion cells corresponds with the lack of type I hair cells in anamniote vertebrates, many of which in amniotes are contacted by the neurites of large, calyx-forming calretinin-immunopositive ganglion cells. In the sensory epithelia of all endorgans, the majority of hair cells were strongly calretinin-immunopositive. Weakly calretinin-immunopositive hair cells were distributed in the intermediate region of the semicircular canal cristae, the central part of the saccular macula, the utricular, and lagenar striola and the medial part of the amphibian papilla. The differential presence of calretinin in the frog vestibular and auditory sensory periphery might reflect a biochemical feature related to firing patterns and frequency bandwidths of self-motion versus acoustic stimulus encoding, respectively.
Collapse
Affiliation(s)
| | | | - Hans Straka
- Department Biology II, Ludwig-Maximilians-University Munich, Planegg, Germany
| |
Collapse
|
43
|
El-Far AH, Elewa YHA, Abdelfattah EZA, Alsenosy AWA, Atta MS, Abou-Zeid KM, Al Jaouni SK, Mousa SA, Noreldin AE. RETRACTED: Thymoquinone and Curcumin Defeat Aging-Associated Oxidative Alterations Induced by D-Galactose in Rats' Brain and Heart. Int J Mol Sci 2021; 22:6839. [PMID: 34202112 PMCID: PMC8268720 DOI: 10.3390/ijms22136839] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/09/2021] [Accepted: 06/18/2021] [Indexed: 01/17/2023] Open
Abstract
D-galactose (D-gal) administration causes oxidative disorder and is widely utilized in aging animal models. Therefore, we subcutaneously injected D-gal at 200 mg/kg BW dose to assess the potential preventive effect of thymoquinone (TQ) and curcumin (Cur) against the oxidative alterations induced by D-gal. Other than the control, vehicle, and D-gal groups, the TQ and Cur treated groups were orally supplemented at 20 mg/kg BW of each alone or combined. TQ and Cur effectively suppressed the oxidative alterations induced by D-gal in brain and heart tissues. The TQ and Cur combination significantly decreased the elevated necrosis in the brain and heart by D-gal. It significantly reduced brain caspase 3, calbindin, and calcium-binding adapter molecule 1 (IBA1), heart caspase 3, and BCL2. Expression of mRNA of the brain and heart TP53, p21, Bax, and CASP-3 were significantly downregulated in the TQ and Cur combination group along with upregulation of BCL2 in comparison with the D-gal group. Data suggested that the TQ and Cur combination is a promising approach in aging prevention.
Collapse
Affiliation(s)
- Ali H. El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt;
| | - Yaser H. A. Elewa
- Department of Histology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
- Laboratory of Anatomy, Faculty of Veterinary Medicine, Basic Veterinary Sciences, Hokkaido University, Sapporo 060-0818, Japan
| | | | - Abdel-Wahab A. Alsenosy
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt;
| | - Mustafa S. Atta
- Department of Physiology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Khalid M. Abou-Zeid
- Animal Care Unit, Medical Research Institute, Alexandria University, Alexandria 21544, Egypt; (E.-Z.A.A.); (K.M.A.-Z.)
| | - Soad K. Al Jaouni
- Department of Hematology/Pediatric Oncology, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Shaker A. Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA;
| | - Ahmed E. Noreldin
- Histology and Cytology Department, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt;
| |
Collapse
|
44
|
Mester JR, Bazzigaluppi P, Dorr A, Beckett T, Burke M, McLaurin J, Sled JG, Stefanovic B. Attenuation of tonic inhibition prevents chronic neurovascular impairments in a Thy1-ChR2 mouse model of repeated, mild traumatic brain injury. Am J Cancer Res 2021; 11:7685-7699. [PMID: 34335958 PMCID: PMC8315057 DOI: 10.7150/thno.60190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/04/2021] [Indexed: 12/24/2022] Open
Abstract
Rationale: Mild traumatic brain injury (mTBI), the most common type of brain trauma, frequently leads to chronic cognitive and neurobehavioral deficits. Intervening effectively is impeded by our poor understanding of its pathophysiological sequelae. Methods: To elucidate the long-term neurovascular sequelae of mTBI, we combined optogenetics, two-photon fluorescence microscopy, and intracortical electrophysiological recordings in mice to selectively stimulate peri-contusional neurons weeks following repeated closed-head injury and probe individual vessel's function and local neuronal reactivity. Results: Compared to sham-operated animals, mTBI mice showed doubled cortical venular speeds (115 ± 25%) and strongly elevated cortical venular reactivity (53 ± 17%). Concomitantly, the pericontusional neurons exhibited attenuated spontaneous activity (-57 ± 79%) and decreased reactivity (-47 ± 28%). Post-mortem immunofluorescence revealed signs of peri-contusional senescence and DNA damage, in the absence of neuronal loss or gliosis. Alteration of neuronal and vascular functioning was largely prevented by chronic, low dose, systemic administration of a GABA-A receptor inverse agonist (L-655,708), commencing 3 days following the third impact. Conclusions: Our findings indicate that repeated mTBI leads to dramatic changes in the neurovascular unit function and that attenuation of tonic inhibition can prevent these alterations. The sustained disruption of the neurovascular function may underlie the concussed brain's long-term susceptibility to injury, and calls for development of better functional assays as well as of neurovascularly targeted interventions.
Collapse
|
45
|
Kelemen K, Szilágyi T. New Approach for Untangling the Role of Uncommon Calcium-Binding Proteins in the Central Nervous System. Brain Sci 2021. [PMID: 34069107 DOI: 10.3390/brainsci11050634ht] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023] Open
Abstract
Although Ca2+ ion plays an essential role in cellular physiology, calcium-binding proteins (CaBPs) were long used for mainly as immunohistochemical markers of specific cell types in different regions of the central nervous system. They are a heterogeneous and wide-ranging group of proteins. Their function was studied intensively in the last two decades and a tremendous amount of information was gathered about them. Girard et al. compiled a comprehensive list of the gene-expression profiles of the entire EF-hand gene superfamily in the murine brain. We selected from this database those CaBPs which are related to information processing and/or neuronal signalling, have a Ca2+-buffer activity, Ca2+-sensor activity, modulator of Ca2+-channel activity, or a yet unknown function. In this way we created a gene function-based selection of the CaBPs. We cross-referenced these findings with publicly available, high-quality RNA-sequencing and in situ hybridization databases (Human Protein Atlas (HPA), Brain RNA-seq database and Allen Brain Atlas integrated into the HPA) and created gene expression heat maps of the regional and cell type-specific expression levels of the selected CaBPs. This represents a useful tool to predict and investigate different expression patterns and functions of the less-known CaBPs of the central nervous system.
Collapse
Affiliation(s)
- Krisztina Kelemen
- Department of Physiology, Doctoral School, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mureș, Romania
| | - Tibor Szilágyi
- Department of Physiology, Doctoral School, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mureș, Romania
| |
Collapse
|
46
|
Kelemen K, Szilágyi T. New Approach for Untangling the Role of Uncommon Calcium-Binding Proteins in the Central Nervous System. Brain Sci 2021; 11:brainsci11050634. [PMID: 34069107 PMCID: PMC8156796 DOI: 10.3390/brainsci11050634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/09/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023] Open
Abstract
Although Ca2+ ion plays an essential role in cellular physiology, calcium-binding proteins (CaBPs) were long used for mainly as immunohistochemical markers of specific cell types in different regions of the central nervous system. They are a heterogeneous and wide-ranging group of proteins. Their function was studied intensively in the last two decades and a tremendous amount of information was gathered about them. Girard et al. compiled a comprehensive list of the gene-expression profiles of the entire EF-hand gene superfamily in the murine brain. We selected from this database those CaBPs which are related to information processing and/or neuronal signalling, have a Ca2+-buffer activity, Ca2+-sensor activity, modulator of Ca2+-channel activity, or a yet unknown function. In this way we created a gene function-based selection of the CaBPs. We cross-referenced these findings with publicly available, high-quality RNA-sequencing and in situ hybridization databases (Human Protein Atlas (HPA), Brain RNA-seq database and Allen Brain Atlas integrated into the HPA) and created gene expression heat maps of the regional and cell type-specific expression levels of the selected CaBPs. This represents a useful tool to predict and investigate different expression patterns and functions of the less-known CaBPs of the central nervous system.
Collapse
|
47
|
Ahamad N, Singh BB. Calcium channels and their role in regenerative medicine. World J Stem Cells 2021; 13:260-280. [PMID: 33959218 PMCID: PMC8080543 DOI: 10.4252/wjsc.v13.i4.260] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/22/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Stem cells hold indefinite self-renewable capability that can be differentiated into all desired cell types. Based on their plasticity potential, they are divided into totipotent (morula stage cells), pluripotent (embryonic stem cells), multipotent (hematopoietic stem cells, multipotent adult progenitor stem cells, and mesenchymal stem cells [MSCs]), and unipotent (progenitor cells that differentiate into a single lineage) cells. Though bone marrow is the primary source of multipotent stem cells in adults, other tissues such as adipose tissues, placenta, amniotic fluid, umbilical cord blood, periodontal ligament, and dental pulp also harbor stem cells that can be used for regenerative therapy. In addition, induced pluripotent stem cells also exhibit fundamental properties of self-renewal and differentiation into specialized cells, and thus could be another source for regenerative medicine. Several diseases including neurodegenerative diseases, cardiovascular diseases, autoimmune diseases, virus infection (also coronavirus disease 2019) have limited success with conventional medicine, and stem cell transplantation is assumed to be the best therapy to treat these disorders. Importantly, MSCs, are by far the best for regenerative medicine due to their limited immune modulation and adequate tissue repair. Moreover, MSCs have the potential to migrate towards the damaged area, which is regulated by various factors and signaling processes. Recent studies have shown that extracellular calcium (Ca2+) promotes the proliferation of MSCs, and thus can assist in transplantation therapy. Ca2+ signaling is a highly adaptable intracellular signal that contains several components such as cell-surface receptors, Ca2+ channels/pumps/exchangers, Ca2+ buffers, and Ca2+ sensors, which together are essential for the appropriate functioning of stem cells and thus modulate their proliferative and regenerative capacity, which will be discussed in this review.
Collapse
Affiliation(s)
- Nassem Ahamad
- School of Dentistry, UT Health Science Center San Antonio, San Antonio, TX 78257, United States
| | - Brij B Singh
- School of Dentistry, UT Health Science Center San Antonio, San Antonio, TX 78257, United States
| |
Collapse
|
48
|
Brain CHID1 Expression Correlates with NRGN and CALB1 in Healthy Subjects and AD Patients. Cells 2021; 10:cells10040882. [PMID: 33924468 PMCID: PMC8069241 DOI: 10.3390/cells10040882] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/06/2021] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer’s disease is a progressive, devastating, and irreversible brain disorder that, day by day, destroys memory skills and social behavior. Despite this, the number of known genes suitable for discriminating between AD patients is insufficient. Among the genes potentially involved in the development of AD, there are the chitinase-like proteins (CLPs) CHI3L1, CHI3L2, and CHID1. The genes of the first two have been extensively investigated while, on the contrary, little information is available on CHID1. In this manuscript, we conducted transcriptome meta-analysis on an extensive sample of brains of healthy control subjects (n = 1849) (NDHC) and brains of AD patients (n = 1170) in order to demonstrate CHID1 involvement. Our analysis revealed an inverse correlation between the brain CHID1 expression levels and the age of NDHC subjects. Significant differences were highlighted comparing CHID1 expression of NDHC subjects and AD patients. Exclusive in AD patients, the CHID1 expression levels were correlated positively to calcium-binding adapter molecule 1 (IBA1) levels. Furthermore, both in NDHC and in AD patient’s brains, the CHID1 expression levels were directly correlated with calbindin 1 (CALB1) and neurogranin (NRGN). According to brain regions, correlation differences were shown between the expression levels of CHID1 in prefrontal, frontal, occipital, cerebellum, temporal, and limbic system. Sex-related differences were only highlighted in NDHC. CHID1 represents a new chitinase potentially involved in the principal processes underlying Alzheimer’s disease.
Collapse
|
49
|
Miguel JC, Perez SE, Malek-Ahmadi M, Mufson EJ. Cerebellar Calcium-Binding Protein and Neurotrophin Receptor Defects in Down Syndrome and Alzheimer's Disease. Front Aging Neurosci 2021; 13:645334. [PMID: 33776745 PMCID: PMC7994928 DOI: 10.3389/fnagi.2021.645334] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/04/2021] [Indexed: 12/11/2022] Open
Abstract
Cerebellar hypoplasia is a major characteristic of the Down syndrome (DS) brain. However, the consequences of trisomy upon cerebellar Purkinje cells (PC) and interneurons in DS are unclear. The present study performed a quantitative and qualitative analysis of cerebellar neurons immunostained with antibodies against calbindin D-28k (Calb), parvalbumin (Parv), and calretinin (Calr), phosphorylated and non-phosphorylated intermediate neurofilaments (SMI-34 and SMI-32), and high (TrkA) and low (p75NTR) affinity nerve growth factor (NGF) receptors as well as tau and amyloid in DS (n = 12), Alzheimer's disease (AD) (n = 10), and healthy non-dementia control (HC) (n = 8) cases. Our findings revealed higher Aβ42 plaque load in DS compared to AD and HC but no differences in APP/Aβ plaque load between HC, AD, and DS. The cerebellar cortex neither displayed Aβ40 containing plaques nor pathologic phosphorylated tau in any of the cases examined. The number and optical density (OD) measurements of Calb immunoreactive (-ir) PC soma and dendrites were similar between groups, while the number of PCs positive for Parv and SMI-32 were significantly reduced in AD and DS compared to HC. By contrast, the number of SMI-34-ir PC dystrophic axonal swellings, termed torpedoes, was significantly greater in AD compared to DS. No differences in SMI-32- and Parv-ir PC OD measurements were observed between groups. Conversely, total number of Parv- (stellate/basket) and Calr (Lugaro, brush, and Golgi)-positive interneurons were significantly reduced in DS compared to AD and HC. A strong negative correlation was found between counts for Parv-ir interneurons, Calr-ir Golgi and brush cells, and Aβ42 plaque load. Number of TrkA and p75NTR positive PCs were reduced in AD compared to HC. These findings suggest that disturbances in calcium binding proteins play a critical role in cerebellar neuronal dysfunction in adults with DS.
Collapse
Affiliation(s)
- Jennifer C. Miguel
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Sylvia E. Perez
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Michael Malek-Ahmadi
- Department of Biomedical Informatics, Banner Alzheimer's Institute, Phoenix, AZ, United States
| | - Elliott J. Mufson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, United States
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ, United States
| |
Collapse
|
50
|
Fiuza FP, Queiroz JPG, Aquino ACQ, Câmara DA, Brandão LEM, Lima RH, Cavalcanti JRLP, Engelberth RCGJ, Cavalcante JS. Aging Alters Daily and Regional Calretinin Neuronal Expression in the Rat Non-image Forming Visual Thalamus. Front Aging Neurosci 2021; 13:613305. [PMID: 33716710 PMCID: PMC7943479 DOI: 10.3389/fnagi.2021.613305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 02/01/2021] [Indexed: 11/29/2022] Open
Abstract
Aging affects the overall physiology, including the image-forming and non-image forming visual systems. Among the components of the latter, the thalamic retinorecipient inter-geniculate leaflet (IGL) and ventral lateral geniculate (vLGN) nucleus conveys light information to subcortical regions, adjusting visuomotor, and circadian functions. It is noteworthy that several visual related cells, such as neuronal subpopulations in the IGL and vLGN are neurochemically characterized by the presence of calcium binding proteins. Calretinin (CR), a representative of such proteins, denotes region-specificity in a temporal manner by variable day–night expression. In parallel, age-related brain dysfunction and neurodegeneration are associated with abnormal intracellular concentrations of calcium. Here, we investigated whether daily changes in the number of CR neurons are a feature of the aged IGL and vLGN in rats. To this end, we perfused rats, ranging from 3 to 24 months of age, within distinct phases of the day, namely zeitgeber times (ZTs). Then, we evaluated CR immunolabeling through design-based stereological cell estimation. We observed distinct daily rhythms of CR expression in the IGL and in both the retinorecipient (vLGNe) and non-retinorecipient (vLGNi) portions of the vLGN. In the ZT 6, the middle of the light phase, the CR cells are reduced with aging in the IGL and vLGNe. In the ZT 12, the transition between light to dark, an age-related CR loss was found in all nuclei. While CR expression predominates in specific spatial domains of vLGN, age-related changes appear not to be restricted at particular portions. No alterations were found in the dark/light transition or in the middle of the dark phase, ZTs 0, and 18, respectively. These results are relevant in the understanding of how aging shifts the phenotype of visual related cells at topographically organized channels of visuomotor and circadian processing.
Collapse
Affiliation(s)
- Felipe P Fiuza
- Graduate Program in Neuroengineering, Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaíba, Brazil
| | - José Pablo G Queiroz
- Graduate Program in Neuroengineering, Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaíba, Brazil
| | - Antônio Carlos Q Aquino
- Laboratory of Neurochemical Studies, Department of Physiology, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Diego A Câmara
- Laboratory of Neurochemical Studies, Department of Physiology, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Luiz Eduardo M Brandão
- Laboratory of Neurochemical Studies, Department of Physiology, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil.,Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Ramon H Lima
- Graduate Program in Neuroengineering, Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Macaíba, Brazil
| | - José Rodolfo L P Cavalcanti
- Laboratory of Experimental Neurology, Department of Biomedical Sciences, Health Science Center, University of State of Rio Grande do Norte, Mossoró, Brazil
| | - Rovena Clara G J Engelberth
- Laboratory of Neurochemical Studies, Department of Physiology, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Jeferson S Cavalcante
- Laboratory of Neurochemical Studies, Department of Physiology, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil
| |
Collapse
|