1
|
Tanaka E, Yamasaki R, Saitoh BY, Abdelhadi A, Nagata S, Yoshidomi S, Inoue Y, Matsumoto K, Kira JI, Isobe N. Postnatal Allergic Inhalation Induces Glial Inflammation in the Olfactory Bulb and Leads to Autism-Like Traits in Mice. Int J Mol Sci 2024; 25:10464. [PMID: 39408806 PMCID: PMC11476352 DOI: 10.3390/ijms251910464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Autism spectrum disorder (ASD) is one of the most prevalent neurodevelopmental disorders. To explore its pathophysiology, we investigated the association between neonatal allergic exposure and behavioral changes. Adult female C57BL/6J mice were immunized with adjuvant (aluminum hydroxide) or ovalbumin emulsified with adjuvant. After immunization, the mice were mated, and offspring were born at full term. The postnatal dams and infants were then simultaneously exposed to an allergen (ovalbumin) or vehicle via inhalation. After weaning, behavioral testing and histopathological analyses were conducted on male offspring. Compared with the vehicle-exposed offspring, the ovalbumin-exposed offspring had decreased sociability and increased repetitive behavior, thus representing an ASD-like phenotype in mice. Moreover, histopathological analyses revealed that the ovalbumin-exposed mice had increased astroglial, microglial, and eosinophilic infiltration in the olfactory bulb, as well as increased eosinophils in the nasal mucosa. The ovalbumin-exposed mice also had decreased dendritic spine density and a lower proportion of mature spines, suggesting the impairment of stimulus-induced synaptogenesis. In conclusion, postnatal allergic exposure induced an ASD-like phenotype, as well as allergic rhinitis, which was followed by glial inflammation in the olfactory bulb parenchyma.
Collapse
Affiliation(s)
- Eizo Tanaka
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- Department of Neurology, Miyazaki Prefectural Miyazaki Hospital, 5-30 Kita-Takamatsu-Cho, Miyazaki 880-8510, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ban-yu Saitoh
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- Department of Neurology, Himeno Hospital, 2316 Oaza-Nishiro, Hirokawa-machi, Yame-gun, Fukuoka 834-0115, Japan
| | - Amina Abdelhadi
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Zagazig University, Zagazig 44519, Al-Sharqia Governorate, Egypt
| | - Satoshi Nagata
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- Clinical Education Center, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Sato Yoshidomi
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yuka Inoue
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Koichiro Matsumoto
- Division of Respirology, Department of Medicine, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
| | - Jun-ichi Kira
- Translational Neuroscience Center, Graduate School of Medicine, and School of Pharmacy at Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Okawa 831-8501, Japan
- Department of Neurology, Brain and Nerve Center, Fukuoka Central Hospital, 2-6-11 Yakuin, Chuo-ku, Fukuoka 810-0022, Japan
| | - Noriko Isobe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
2
|
Svandova E, Vesela B, Janeckova E, Chai Y, Matalova E. Exploring caspase functions in mouse models. Apoptosis 2024; 29:938-966. [PMID: 38824481 PMCID: PMC11263464 DOI: 10.1007/s10495-024-01976-z] [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] [Accepted: 05/02/2024] [Indexed: 06/03/2024]
Abstract
Caspases are enzymes with protease activity. Despite being known for more than three decades, caspase investigation still yields surprising and fascinating information. Initially associated with cell death and inflammation, their functions have gradually been revealed to extend beyond, targeting pathways such as cell proliferation, migration, and differentiation. These processes are also associated with disease mechanisms, positioning caspases as potential targets for numerous pathologies including inflammatory, neurological, metabolic, or oncological conditions. While in vitro studies play a crucial role in elucidating molecular pathways, they lack the context of the body's complexity. Therefore, laboratory animals are an indispensable part of successfully understanding and applying caspase networks. This paper aims to summarize and discuss recent knowledge, understanding, and challenges in caspase knock-out mice.
Collapse
Affiliation(s)
- Eva Svandova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetic, Brno, Czech Republic.
| | - Barbora Vesela
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetic, Brno, Czech Republic
| | - Eva Janeckova
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, USA
| | - Eva Matalova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetic, Brno, Czech Republic
- Department of Physiology, University of Veterinary Sciences, Brno, Czech Republic
| |
Collapse
|
3
|
Yamaguchi Y, Okamura K, Yamamuro K, Okumura K, Komori T, Toritsuka M, Takada R, Nishihata Y, Ikawa D, Yamauchi T, Makinodan M, Yoshino H, Saito Y, Matsuzaki H, Kishimoto T, Kimoto S. NARP-related alterations in the excitatory and inhibitory circuitry of socially isolated mice: developmental insights and implications for autism spectrum disorder. Front Psychiatry 2024; 15:1403476. [PMID: 38903649 PMCID: PMC11187327 DOI: 10.3389/fpsyt.2024.1403476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/17/2024] [Indexed: 06/22/2024] Open
Abstract
Background Social isolation during critical periods of development is associated with alterations in behavior and neuronal circuitry. This study aimed to investigate the immediate and developmental effects of social isolation on firing properties, neuronal activity-regulated pentraxin (NARP) and parvalbumin (PV) expression in the prefrontal cortex (PFC), social behavior in juvenile socially isolated mice, and the biological relevance of NARP expression in autism spectrum disorder (ASD). Methods Mice were subjected to social isolation during postnatal days 21-35 (P21-P35) and were compared with group-housed control mice. Firing properties in the PFC pyramidal neurons were altered in P35 socially isolated mice, which might be associated with alterations in NARP and PV expression. Results In adulthood, mice that underwent juvenile social isolation exhibited difficulty distinguishing between novel and familiar mice during a social memory task, while maintaining similar levels of social interaction as the control mice. Furthermore, a marked decrease in NARP expression in lymphoblastoid cell lines derived from adolescent humans with ASD as compared to typically developing (TD) humans was found. Conclusion Our study highlights the role of electrophysiological properties, as well as NARP and PV expression in the PFC in mediating the developmental consequences of social isolation on behavior.
Collapse
Affiliation(s)
- Yasunari Yamaguchi
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
- Department of Neuropsychiatry, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Kazuya Okamura
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
- Department of Neuropsychiatry, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Kazuhiko Yamamuro
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Kazuki Okumura
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Takashi Komori
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Michihiro Toritsuka
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Ryohei Takada
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Yosuke Nishihata
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Daisuke Ikawa
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Takahira Yamauchi
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Manabu Makinodan
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Hiroki Yoshino
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
- Mie Prefectural Mental Medical Center, Mie, Japan
| | - Yasuhiko Saito
- Department of Neurophysiology, Nara Medical University School of Medicine, Nara, Japan
| | - Hideo Matsuzaki
- Research Center for Child Mental Development, University of Fukui, Fukui, Japan
| | - Toshifumi Kishimoto
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Sohei Kimoto
- Department of Neuropsychiatry, Wakayama Medical University School of Medicine, Wakayama, Japan
| |
Collapse
|
4
|
Almishri W, Altonsy MO, Swain MG. Cholestatic liver disease leads to significant adaptative changes in neural circuits regulating social behavior in mice to enhance sociability. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167100. [PMID: 38412926 DOI: 10.1016/j.bbadis.2024.167100] [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: 10/16/2023] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND & AIMS Cholestatic liver diseases (CLD) are commonly associated with behavioral changes, including social isolation, that negatively affects patient quality of life and remains unaltered by current therapies. It remains unclear whether CLD-associated social dysfunction stems from a direct effect on the brain, or from the psychological impact of CLD. The psychological component of disease is absent in animals, so we investigated the impact of CLD on social behavior and gene expression profiles in key social behavior-regulating brain regions in a mouse model. METHODS CLD due to bile duct ligation was used with the three-chamber sociability test for behavioral phenotyping. Differentially expressed gene (DEG) signatures were delineated in 3 key brain regions regulating social behavior using RNA-seq. Ingenuity Pathway Analysis (IPA®) was applied to streamline DEG data interpretation and integrate findings with social behavior-regulating pathways to identify important brain molecular networks and regulatory mechanisms disrupted in CLD. RESULTS CLD mice exhibited enhanced social interactive behavior and significantly altered gene expression in each of the three social behavior-regulating brain regions examined. DEG signatures in BDL mice were associated with key IPA®-identified social behavior-regulating pathways including Oxytocin in Brain Signaling, GABA Receptor Signaling, Dopamine Receptor Signaling, and Glutamate Receptor Signaling. CONCLUSIONS CLD causes complex alterations in gene expression profiles in key social behavior-regulating brain areas/pathways linked to enhanced social interactive behavior. These findings, if paralleled in CLD patients, suggest that CLD-associated reductions in social interactions predominantly relate to psychological impacts of disease and may inform new approaches to improve management.
Collapse
Affiliation(s)
- Wagdi Almishri
- Department of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Mohammed O Altonsy
- Department of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada; Department of Zoology, Faculty of Science, Sohag University, Sohag, Egypt
| | - Mark G Swain
- Department of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada; University of Calgary Liver Unit, Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
5
|
Berroug L, Laaroussi M, Essaidi O, Malqui H, Anarghou H, Chaoui AA, Najimi M, Chigr F. Sex-specific neurobehavioral and biochemical effects of developmental exposure to Malathion in offspring mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2215-2231. [PMID: 37804342 DOI: 10.1007/s00210-023-02749-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/25/2023] [Indexed: 10/09/2023]
Abstract
Malathion is an organophosphate pesticide (OP) commonly used in agriculture, industry, and veterinary medicine. Sex is a crucial factor in responding to neurotoxicants, yet the sex-specific effects of OP exposure, particularly neurological impairments following chronic low-level exposure remains limited. Our study aims to evaluate the neurobehavioral and biochemical effects of developmental exposure to Malathion across sexes. Pregnant mice were exposed to a low oral dose of Malathion from gestation up to the weaning of the pups, which were individually gavaged with a similar dose regimen until postnatal day 70. Our results show that Malathion decreased body weight and food intake, reduced locomotor activity and recognition memory. Motor coordination and special memory were only altered in females, whereas we found a male-specific effect of Malathion on social behavior and marble burying. These alterations were accompanied by increased malondialdehyde (MDA), decreased brain acetylcholinesterase activity (AChE), and disrupted brain redox homeostasis. Our findings about the effects of Malathion exposure across sexes may, in part, contribute to understanding the dimorphic susceptibilities observed in neurological disorders.
Collapse
Affiliation(s)
- Laila Berroug
- Biological Engineering Laboratory, Faculty of Science and Technology, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Meriem Laaroussi
- Biological Engineering Laboratory, Faculty of Science and Technology, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Oumaima Essaidi
- Biological Engineering Laboratory, Faculty of Science and Technology, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Hafsa Malqui
- Biological Engineering Laboratory, Faculty of Science and Technology, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Hammou Anarghou
- Biological Engineering Laboratory, Faculty of Science and Technology, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Ahmed Ait Chaoui
- Biological Engineering Laboratory, Faculty of Science and Technology, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Mohamed Najimi
- Biological Engineering Laboratory, Faculty of Science and Technology, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Fatiha Chigr
- Biological Engineering Laboratory, Faculty of Science and Technology, Sultan Moulay Slimane University, Beni Mellal, Morocco.
| |
Collapse
|
6
|
Buchenauer L, Haange SB, Bauer M, Rolle-Kampczyk UE, Wagner M, Stucke J, Elter E, Fink B, Vass M, von Bergen M, Schulz A, Zenclussen AC, Junge KM, Stangl GI, Polte T. Maternal exposure of mice to glyphosate induces depression- and anxiety-like behavior in the offspring via alterations of the gut-brain axis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 905:167034. [PMID: 37709081 DOI: 10.1016/j.scitotenv.2023.167034] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/24/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023]
Abstract
The past decade has been characterized by increased awareness and de-stigmatization of mental health issues, in particular the most common neuropsychiatric disorders depression and anxiety. Further, with growing understanding of neurodevelopmental disorders such as attention deficit and hyperactivity disorder and autism spectrum disorder, the number of diagnosed patients has increased. The pathogenesis of these behavioral disorders is multifactorial and early-life exposure to environmental chemicals has been proposed to be a relevant risk factor that might mediate these effects by disturbances on the gut-brain-axis. However, for glyphosate, the most widely used pesticide worldwide, there are only limited and inconsistent findings that link chronic low-dose exposure in particular during early life to neurobehavioral disorders. Here, we explored the impact of maternal oral glyphosate exposure (0.5 and 50 mg/kg body weight/day) during pregnancy and the lactational period on offspring's behavior, brain gene expression and gut microbiota using a cross-generational mouse model. Behavioral analyses revealed a depression- and anxiety-like behavior as well as social deficits most notably in adult female offspring of glyphosate-exposed dams. Furthermore, the expression of tryptophan hydroxylase 2, an enzyme discussed to be linked to behavioral problems, was reduced in the hippocampus of female offspring and correlated to a glyphosate-induced DNA hypermethylation of the gene. Moreover, maternal glyphosate exposure significantly altered the gut microbiota in the female offspring including a decreased abundance of Akkermansia and increased abundance of Alistipes and Blautia, bacteria involved in tryptophan metabolism and associated with depression- and anxiety-like disorders. Our results suggest that glyphosate might influence the gut-brain axis crosstalk following in-utero and lactational exposure. This study underlines the importance of understanding the impact of exposure to pesticides on the gut-brain axis and further emphasizes the need for microbiome analyses to be compulsorily included in health risk assessments of pesticides.
Collapse
Affiliation(s)
- Lisa Buchenauer
- Helmholtz Centre for Environmental Research - UFZ, Department of Environmental Immunology, Leipzig, Germany; University of Leipzig, Leipzig University Medical Center, Department of Dermatology, Venerology and Allergology, Leipzig, Germany
| | - Sven-Bastiaan Haange
- Helmholtz Centre for Environmental Research - UFZ, Department of Molecular Systems Biology, Leipzig, Germany
| | - Mario Bauer
- Helmholtz Centre for Environmental Research - UFZ, Department of Environmental Immunology, Leipzig, Germany
| | - Ulrike E Rolle-Kampczyk
- Helmholtz Centre for Environmental Research - UFZ, Department of Molecular Systems Biology, Leipzig, Germany
| | - Marita Wagner
- Helmholtz Centre for Environmental Research - UFZ, Department of Environmental Immunology, Leipzig, Germany; University of Leipzig, Leipzig University Medical Center, Department of Dermatology, Venerology and Allergology, Leipzig, Germany
| | - Johanna Stucke
- Helmholtz Centre for Environmental Research - UFZ, Department of Environmental Immunology, Leipzig, Germany; University of Leipzig, Leipzig University Medical Center, Department of Dermatology, Venerology and Allergology, Leipzig, Germany
| | - Elena Elter
- Helmholtz Centre for Environmental Research - UFZ, Department of Environmental Immunology, Leipzig, Germany; University of Leipzig, Leipzig University Medical Center, Department of Dermatology, Venerology and Allergology, Leipzig, Germany
| | - Beate Fink
- Helmholtz Centre for Environmental Research - UFZ, Department of Environmental Immunology, Leipzig, Germany
| | - Maren Vass
- University of Leipzig, Leipzig University Medical Center, Department of Dermatology, Venerology and Allergology, Leipzig, Germany
| | - Martin von Bergen
- Helmholtz Centre for Environmental Research - UFZ, Department of Molecular Systems Biology, Leipzig, Germany; University of Leipzig, Faculty of Life Sciences, Institute of Biochemistry, Leipzig, Germany; German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
| | - Angela Schulz
- University of Leipzig, Medical Faculty, Rudolf Schönheimer Institute of Biochemistry, Leipzig, Germany
| | - Ana C Zenclussen
- Helmholtz Centre for Environmental Research - UFZ, Department of Environmental Immunology, Leipzig, Germany; Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, University Leipzig, 04103 Leipzig, Germany
| | - Kristin M Junge
- Helmholtz Centre for Environmental Research - UFZ, Department of Environmental Immunology, Leipzig, Germany; AKAD University Stuttgart, School of Health and Social Sciences, Stuttgart, Germany
| | - Gabriele I Stangl
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Tobias Polte
- Helmholtz Centre for Environmental Research - UFZ, Department of Environmental Immunology, Leipzig, Germany; University of Leipzig, Leipzig University Medical Center, Department of Dermatology, Venerology and Allergology, Leipzig, Germany.
| |
Collapse
|
7
|
Arakawa H. Revisiting sociability: Factors facilitating approach and avoidance during the three-chamber test. Physiol Behav 2023; 272:114373. [PMID: 37805136 DOI: 10.1016/j.physbeh.2023.114373] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/07/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
The three-chamber test, the so-called sociability test, has been widely used to assess social deficits based on impaired socially oriented investigations in rodent models. An innate motivation for investigating conspecifics is theoretically a prerequisite for gaining sociability scores in this paradigm. However, several relevant factors mediating investigatory motives, such as familiarity, attractiveness, and aggression, may affect sociability scores, which must be verified to obtain an adequate evaluation of the psychiatric phenotypes exhibited by disease-relevant rodent models. We assessed the social and non-social factors that mediate proximity preference by the three-chamber test with standard C57BL/6 J (B6) mice and low sociability BTBR+ltpr3tf/J (BTBR) mice. Strains of the opponents had no effect. Sexual cues (i.e., opposite sex) increased proximity preference in both strains of mice; in contrast, novel objects induced an approach in B6 mice but avoidance in BTBR mice. Single-housing before testing, stimulated social motive, affected BTBR mice but not B6 mice. BTBR females showed increased proximity preference across the sessions, and BTBR males showed increased preference toward a male B6 stimulus, but not a male BTBR stimulus. The male preference was restored when the male BTBR stimulus was anesthetized. In addition, self-grooming was facilitated by social and non-social novelty cues in both strains. B6 mice predominantly exhibited an investigatory approach toward social or non-social stimuli, whereas BTBR mice recognized social cues but tended to show avoidance. The three-chamber test could evaluate approach-avoidance strategies in target mouse strains that comprise innate social distance between mice.
Collapse
Affiliation(s)
- Hiroyuki Arakawa
- Department Systems Physiology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa 903-0215, Japan.
| |
Collapse
|
8
|
Abdelrazik E, Hassan HM, Hamza E, Ezz Elregal FM, Elnagdy MH, Abdulhai EA. Beneficial role of rosemary extract on oxidative stress-mediated neuronal apoptosis in rotenone-induced attention deficit hyperactivity disease in juvenile rat model. ACTA BIO-MEDICA : ATENEI PARMENSIS 2023; 94:e2023104. [PMID: 37326266 PMCID: PMC10308472 DOI: 10.23750/abm.v94i3.14260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 02/23/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND AND AIM Attention deficit hyperactivity disorder (ADHD) is heterogeneous neurobehavioral disorders that co-exist with cognitive and learning deficits affecting 3-7% of children. We study the role of rosemary in the protection of the prefrontal cortical neurons against rotenone-induced ADHD in juvenile rats. METHODS Twenty-four juvenile rats were divided into four groups (n=6): control group, received olive oil 0.5 ml/kg/day/ I.P. for 4 weeks, rosemary group received rosemary 75 mg/kg/day/ I.P. for 4 weeks, rotenone group received rotenone 1 mg/kg/day/ I.P. dissolved in olive oil for 4 days and combined group received rotenone 1 mg/kg/day/ I.P. for 4 days and rosemary 75 mg/kg/day/ I.P. for 4 weeks. RESULTS Rotenone group showed higher impulsivity with reduction in the recognition index and total locomotor activity. However, combined group showed significant improvement in the recognition index and the total locomotor activity. Neurochemical analysis disclosed that rotenone decreased levels of GSH and significantly increased lipid peroxidation and oxidative stress. The administration of rosemary amended these neurochemical changes. Rotenone caused a significant increase in serum amyloid protein A and C-reactive protein levels indicating a marked state of inflammation. Rosemary ameliorated these biochemical changes. The immunohistochemical expression of tyrosine hydroxylase was decreased in the rotenone group. On the other hand, caspase-3 was increased in the rotenone group. PCR confirmed immunohistochemical results for gene expression. CONCLUSIONS The findings of the behavioral, neurochemical, biochemical, immunohistochemical and molecular outcomes suggested that rosemary could fight oxidative stress, inflammation and apoptosis in the prefrontal cortex of rotenone-induced ADHD in juvenile rats.
Collapse
Affiliation(s)
- Eman Abdelrazik
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt. .
| | - Hend M Hassan
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt. .
| | - Eman Hamza
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt/ Department of Biochemistry and Molecular Biology, Horus University, Damietta, Egypt..
| | - Farah M Ezz Elregal
- Medical Student, Faculty of Medicine, Mansoura University, Mansoura, Egypt. .
| | - Marwa H Elnagdy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt..
| | - Eman A Abdulhai
- Department of Pediatrics (pediatric neurology), Faculty of Medicine, Mansoura University, Mansoura, Egypt. .
| |
Collapse
|
9
|
Vitale I, Pietrocola F, Guilbaud E, Aaronson SA, Abrams JM, Adam D, Agostini M, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, Aqeilan RI, Arama E, Baehrecke EH, Balachandran S, Bano D, Barlev NA, Bartek J, Bazan NG, Becker C, Bernassola F, Bertrand MJM, Bianchi ME, Blagosklonny MV, Blander JM, Blandino G, Blomgren K, Borner C, Bortner CD, Bove P, Boya P, Brenner C, Broz P, Brunner T, Damgaard RB, Calin GA, Campanella M, Candi E, Carbone M, Carmona-Gutierrez D, Cecconi F, Chan FKM, Chen GQ, Chen Q, Chen YH, Cheng EH, Chipuk JE, Cidlowski JA, Ciechanover A, Ciliberto G, Conrad M, Cubillos-Ruiz JR, Czabotar PE, D'Angiolella V, Daugaard M, Dawson TM, Dawson VL, De Maria R, De Strooper B, Debatin KM, Deberardinis RJ, Degterev A, Del Sal G, Deshmukh M, Di Virgilio F, Diederich M, Dixon SJ, Dynlacht BD, El-Deiry WS, Elrod JW, Engeland K, Fimia GM, Galassi C, Ganini C, Garcia-Saez AJ, Garg AD, Garrido C, Gavathiotis E, Gerlic M, Ghosh S, Green DR, Greene LA, Gronemeyer H, Häcker G, Hajnóczky G, Hardwick JM, Haupt Y, He S, Heery DM, Hengartner MO, Hetz C, Hildeman DA, Ichijo H, Inoue S, Jäättelä M, Janic A, Joseph B, Jost PJ, Kanneganti TD, Karin M, Kashkar H, Kaufmann T, Kelly GL, Kepp O, Kimchi A, Kitsis RN, Klionsky DJ, Kluck R, Krysko DV, Kulms D, Kumar S, Lavandero S, Lavrik IN, Lemasters JJ, Liccardi G, Linkermann A, Lipton SA, Lockshin RA, López-Otín C, Luedde T, MacFarlane M, Madeo F, Malorni W, Manic G, Mantovani R, Marchi S, Marine JC, Martin SJ, Martinou JC, Mastroberardino PG, Medema JP, Mehlen P, Meier P, Melino G, Melino S, Miao EA, Moll UM, Muñoz-Pinedo C, Murphy DJ, Niklison-Chirou MV, Novelli F, Núñez G, Oberst A, Ofengeim D, Opferman JT, Oren M, Pagano M, Panaretakis T, Pasparakis M, Penninger JM, Pentimalli F, Pereira DM, Pervaiz S, Peter ME, Pinton P, Porta G, Prehn JHM, Puthalakath H, Rabinovich GA, Rajalingam K, Ravichandran KS, Rehm M, Ricci JE, Rizzuto R, Robinson N, Rodrigues CMP, Rotblat B, Rothlin CV, Rubinsztein DC, Rudel T, Rufini A, Ryan KM, Sarosiek KA, Sawa A, Sayan E, Schroder K, Scorrano L, Sesti F, Shao F, Shi Y, Sica GS, Silke J, Simon HU, Sistigu A, Stephanou A, Stockwell BR, Strapazzon F, Strasser A, Sun L, Sun E, Sun Q, Szabadkai G, Tait SWG, Tang D, Tavernarakis N, Troy CM, Turk B, Urbano N, Vandenabeele P, Vanden Berghe T, Vander Heiden MG, Vanderluit JL, Verkhratsky A, Villunger A, von Karstedt S, Voss AK, Vousden KH, Vucic D, Vuri D, Wagner EF, Walczak H, Wallach D, Wang R, Wang Y, Weber A, Wood W, Yamazaki T, Yang HT, Zakeri Z, Zawacka-Pankau JE, Zhang L, Zhang H, Zhivotovsky B, Zhou W, Piacentini M, Kroemer G, Galluzzi L. Apoptotic cell death in disease-Current understanding of the NCCD 2023. Cell Death Differ 2023; 30:1097-1154. [PMID: 37100955 PMCID: PMC10130819 DOI: 10.1038/s41418-023-01153-w] [Citation(s) in RCA: 112] [Impact Index Per Article: 112.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 04/28/2023] Open
Abstract
Apoptosis is a form of regulated cell death (RCD) that involves proteases of the caspase family. Pharmacological and genetic strategies that experimentally inhibit or delay apoptosis in mammalian systems have elucidated the key contribution of this process not only to (post-)embryonic development and adult tissue homeostasis, but also to the etiology of multiple human disorders. Consistent with this notion, while defects in the molecular machinery for apoptotic cell death impair organismal development and promote oncogenesis, the unwarranted activation of apoptosis promotes cell loss and tissue damage in the context of various neurological, cardiovascular, renal, hepatic, infectious, neoplastic and inflammatory conditions. Here, the Nomenclature Committee on Cell Death (NCCD) gathered to critically summarize an abundant pre-clinical literature mechanistically linking the core apoptotic apparatus to organismal homeostasis in the context of disease.
Collapse
Affiliation(s)
- Ilio Vitale
- IIGM - Italian Institute for Genomic Medicine, c/o IRCSS Candiolo, Torino, Italy.
- Candiolo Cancer Institute, FPO -IRCCS, Candiolo, Italy.
| | - Federico Pietrocola
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Emma Guilbaud
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Stuart A Aaronson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - John M Abrams
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dieter Adam
- Institut für Immunologie, Kiel University, Kiel, Germany
| | - Massimiliano Agostini
- Department of Experimental Medicine, University of Rome Tor Vergata, TOR, Rome, Italy
| | - Patrizia Agostinis
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Emad S Alnemri
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
- BIOGEM, Avellino, Italy
| | - Ivano Amelio
- Division of Systems Toxicology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - David W Andrews
- Sunnybrook Research Institute, Toronto, ON, Canada
- Departments of Biochemistry and Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Rami I Aqeilan
- Hebrew University of Jerusalem, Lautenberg Center for Immunology & Cancer Research, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Jerusalem, Israel
| | - Eli Arama
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eric H Baehrecke
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Siddharth Balachandran
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Daniele Bano
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Nickolai A Barlev
- Department of Biomedicine, Nazarbayev University School of Medicine, Astana, Kazakhstan
| | - Jiri Bartek
- Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, USA
| | - Christoph Becker
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Francesca Bernassola
- Department of Experimental Medicine, University of Rome Tor Vergata, TOR, Rome, Italy
| | - Mathieu J M Bertrand
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Marco E Bianchi
- Università Vita-Salute San Raffaele, School of Medicine, Milan, Italy and Ospedale San Raffaele IRCSS, Milan, Italy
| | | | - J Magarian Blander
- Department of Medicine, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | | | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
- Pediatric Hematology and Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Christoph Borner
- Institute of Molecular Medicine and Cell Research, Medical Faculty, Albert Ludwigs University of Freiburg, Freiburg, Germany
| | - Carl D Bortner
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, Durham, NC, USA
| | - Pierluigi Bove
- Department of Experimental Medicine, University of Rome Tor Vergata, TOR, Rome, Italy
| | - Patricia Boya
- Centro de Investigaciones Biologicas Margarita Salas, CSIC, Madrid, Spain
| | - Catherine Brenner
- Université Paris-Saclay, CNRS, Institut Gustave Roussy, Aspects métaboliques et systémiques de l'oncogénèse pour de nouvelles approches thérapeutiques, Villejuif, France
| | - Petr Broz
- Department of Immunobiology, University of Lausanne, Epalinges, Vaud, Switzerland
| | - Thomas Brunner
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Rune Busk Damgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michelangelo Campanella
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, London, UK
- UCL Consortium for Mitochondrial Research, London, UK
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Eleonora Candi
- Department of Experimental Medicine, University of Rome Tor Vergata, TOR, Rome, Italy
| | - Michele Carbone
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI, USA
| | | | - Francesco Cecconi
- Cell Stress and Survival Unit, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, Copenhagen, Denmark
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francis K-M Chan
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Guo-Qiang Chen
- State Key Lab of Oncogene and its related gene, Ren-Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Quan Chen
- College of Life Sciences, Nankai University, Tianjin, China
| | - Youhai H Chen
- Shenzhen Institute of Advanced Technology (SIAT), Shenzhen, Guangdong, China
| | - Emily H Cheng
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jerry E Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John A Cidlowski
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, Durham, NC, USA
| | - Aaron Ciechanover
- The Technion-Integrated Cancer Center, The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | | | - Marcus Conrad
- Helmholtz Munich, Institute of Metabolism and Cell Death, Neuherberg, Germany
| | - Juan R Cubillos-Ruiz
- Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA
| | - Peter E Czabotar
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | | | - Mads Daugaard
- Department of Urologic Sciences, Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Ted M Dawson
- Institute for Cell Engineering and the Departments of Neurology, Neuroscience and Pharmacology & Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Valina L Dawson
- Institute for Cell Engineering and the Departments of Neurology, Neuroscience and Pharmacology & Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ruggero De Maria
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Bart De Strooper
- VIB Centre for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- The Francis Crick Institute, London, UK
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Ralph J Deberardinis
- Howard Hughes Medical Institute and Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alexei Degterev
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | - Giannino Del Sal
- Department of Life Sciences, University of Trieste, Trieste, Italy
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park-Padriciano, Trieste, Italy
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Mohanish Deshmukh
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | | | - Marc Diederich
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Brian D Dynlacht
- Department of Pathology, New York University Cancer Institute, New York University School of Medicine, New York, NY, USA
| | - Wafik S El-Deiry
- Division of Hematology/Oncology, Brown University and the Lifespan Cancer Institute, Providence, RI, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI, USA
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - John W Elrod
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Kurt Engeland
- Molecular Oncology, University of Leipzig, Leipzig, Germany
| | - Gian Maria Fimia
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases 'L. Spallanzani' IRCCS, Rome, Italy
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Claudia Galassi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Carlo Ganini
- Department of Experimental Medicine, University of Rome Tor Vergata, TOR, Rome, Italy
- Biochemistry Laboratory, Dermopatic Institute of Immaculate (IDI) IRCCS, Rome, Italy
| | - Ana J Garcia-Saez
- CECAD, Institute of Genetics, University of Cologne, Cologne, Germany
| | - Abhishek D Garg
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Carmen Garrido
- INSERM, UMR, 1231, Dijon, France
- Faculty of Medicine, Université de Bourgogne Franche-Comté, Dijon, France
- Anti-cancer Center Georges-François Leclerc, Dijon, France
| | - Evripidis Gavathiotis
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, NY, USA
- Institute for Aging Research, Albert Einstein College of Medicine, New York, NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY, USA
| | - Motti Gerlic
- Department of Clinical Microbiology and Immunology, Sackler school of Medicine, Tel Aviv university, Tel Aviv, Israel
| | - Sourav Ghosh
- Department of Neurology and Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Douglas R Green
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Lloyd A Greene
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Hinrich Gronemeyer
- Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Georg Häcker
- Faculty of Medicine, Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - György Hajnóczky
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - J Marie Hardwick
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Departments of Molecular Microbiology and Immunology, Pharmacology, Oncology and Neurology, Johns Hopkins Bloomberg School of Public Health and School of Medicine, Baltimore, MD, USA
| | - Ygal Haupt
- VITTAIL Ltd, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Sudan He
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, China
| | - David M Heery
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | | | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
- Center for Molecular Studies of the Cell, Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Buck Institute for Research on Aging, Novato, CA, USA
| | - David A Hildeman
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, The University of Tokyo, Tokyo, Japan
| | - Satoshi Inoue
- National Cancer Center Research Institute, Tokyo, Japan
| | - Marja Jäättelä
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ana Janic
- Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona, Spain
| | - Bertrand Joseph
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Philipp J Jost
- Clinical Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | | | - Michael Karin
- Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego, San Diego, CA, USA
| | - Hamid Kashkar
- CECAD Research Center, Institute for Molecular Immunology, University of Cologne, Cologne, Germany
| | - Thomas Kaufmann
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Adi Kimchi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Richard N Kitsis
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, NY, USA
- Institute for Aging Research, Albert Einstein College of Medicine, New York, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, New York, NY, USA
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York, NY, USA
| | | | - Ruth Kluck
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Dmitri V Krysko
- Cell Death Investigation and Therapy Lab, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Dagmar Kulms
- Department of Dermatology, Experimental Dermatology, TU-Dresden, Dresden, Germany
- National Center for Tumor Diseases Dresden, TU-Dresden, Dresden, Germany
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, Australia
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Sergio Lavandero
- Universidad de Chile, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Inna N Lavrik
- Translational Inflammation Research, Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| | - John J Lemasters
- Departments of Drug Discovery & Biomedical Sciences and Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Gianmaria Liccardi
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine 3, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Stuart A Lipton
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Richard A Lockshin
- Department of Biology, Queens College of the City University of New York, Flushing, NY, USA
- St. John's University, Jamaica, NY, USA
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Heinrich Heine University, Duesseldorf, Germany
| | - Marion MacFarlane
- Medical Research Council Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Field of Excellence BioHealth - University of Graz, Graz, Austria
| | - Walter Malorni
- Center for Global Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gwenola Manic
- IIGM - Italian Institute for Genomic Medicine, c/o IRCSS Candiolo, Torino, Italy
- Candiolo Cancer Institute, FPO -IRCCS, Candiolo, Italy
| | - Roberto Mantovani
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| | - Saverio Marchi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Jean-Christophe Marine
- VIB Center for Cancer Biology, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - Jean-Claude Martinou
- Department of Cell Biology, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Pier G Mastroberardino
- Department of Molecular Genetics, Rotterdam, the Netherlands
- IFOM-ETS The AIRC Institute for Molecular Oncology, Milan, Italy
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Jan Paul Medema
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Patrick Mehlen
- Apoptosis, Cancer, and Development Laboratory, Equipe labellisée 'La Ligue', LabEx DEVweCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon1, Lyon, France
| | - Pascal Meier
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Gerry Melino
- Department of Experimental Medicine, University of Rome Tor Vergata, TOR, Rome, Italy
| | - Sonia Melino
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Rome, Italy
| | - Edward A Miao
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Ute M Moll
- Department of Pathology and Stony Brook Cancer Center, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Cristina Muñoz-Pinedo
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Spain
| | - Daniel J Murphy
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Glasgow, UK
| | | | - Flavia Novelli
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Gabriel Núñez
- Department of Pathology and Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
| | - Andrew Oberst
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Dimitry Ofengeim
- Rare and Neuroscience Therapeutic Area, Sanofi, Cambridge, MA, USA
| | - Joseph T Opferman
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Moshe Oren
- Department of Molecular Cell Biology, The Weizmann Institute, Rehovot, Israel
| | - Michele Pagano
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine and Howard Hughes Medical Institute, New York, NY, USA
| | - Theocharis Panaretakis
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of GU Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Josef M Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | | | - David M Pereira
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Shazib Pervaiz
- Department of Physiology, YLL School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore, Singapore
- National University Cancer Institute, NUHS, Singapore, Singapore
- ISEP, NUS Graduate School, National University of Singapore, Singapore, Singapore
| | - Marcus E Peter
- Department of Medicine, Division Hematology/Oncology, Northwestern University, Chicago, IL, USA
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Giovanni Porta
- Center of Genomic Medicine, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin 2, Ireland
| | - Hamsa Puthalakath
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Gabriel A Rabinovich
- Laboratorio de Glicomedicina. Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | | | - Kodi S Ravichandran
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Cell Clearance, Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Markus Rehm
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Jean-Ehrland Ricci
- Université Côte d'Azur, INSERM, C3M, Equipe labellisée Ligue Contre le Cancer, Nice, France
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Nirmal Robinson
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, Australia
| | - Cecilia M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Barak Rotblat
- Department of Life sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
- The NIBN, Beer Sheva, Israel
| | - Carla V Rothlin
- Department of Immunobiology and Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK
| | - Thomas Rudel
- Microbiology Biocentre, University of Würzburg, Würzburg, Germany
| | - Alessandro Rufini
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
- University of Leicester, Leicester Cancer Research Centre, Leicester, UK
| | - Kevin M Ryan
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Kristopher A Sarosiek
- John B. Little Center for Radiation Sciences, Harvard School of Public Health, Boston, MA, USA
- Department of Systems Biology, Lab of Systems Pharmacology, Harvard Program in Therapeutics Science, Harvard Medical School, Boston, MA, USA
- Department of Environmental Health, Molecular and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston, MA, USA
| | - Akira Sawa
- Johns Hopkins Schizophrenia Center, Johns Hopkins University, Baltimore, MD, USA
| | - Emre Sayan
- Faculty of Medicine, Cancer Sciences Unit, University of Southampton, Southampton, UK
| | - Kate Schroder
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Luca Scorrano
- Department of Biology, University of Padua, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| | - Federico Sesti
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, NJ, USA
| | - Feng Shao
- National Institute of Biological Sciences, Beijing, PR China
| | - Yufang Shi
- Department of Experimental Medicine, University of Rome Tor Vergata, TOR, Rome, Italy
- The Third Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, Jiangsu, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Giuseppe S Sica
- Department of Surgical Science, University Tor Vergata, Rome, Italy
| | - John Silke
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany
| | - Antonella Sistigu
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Brent R Stockwell
- Department of Biological Sciences and Department of Chemistry, Columbia University, New York, NY, USA
| | - Flavie Strapazzon
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Univ Lyon, Univ Lyon 1, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyogène CNRS, INSERM, Lyon, France
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Liming Sun
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Erwei Sun
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Qiang Sun
- Laboratory of Cell Engineering, Institute of Biotechnology, Beijing, China
- Research Unit of Cell Death Mechanism, 2021RU008, Chinese Academy of Medical Science, Beijing, China
| | - Gyorgy Szabadkai
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, UK
| | - Stephen W G Tait
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Daolin Tang
- Department of Surgery, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
- Department of Basic Sciences, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Carol M Troy
- Departments of Pathology & Cell Biology and Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, J. Stefan Institute, Ljubljana, Slovenia
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Nicoletta Urbano
- Department of Oncohaematology, University of Rome Tor Vergata, TOR, Rome, Italy
| | - Peter Vandenabeele
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Methusalem Program, Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Achucarro Center for Neuroscience, IKERBASQUE, Bilbao, Spain
- School of Forensic Medicine, China Medical University, Shenyang, China
- State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Andreas Villunger
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
- The Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences (OeAW), Vienna, Austria
- The Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria
| | - Silvia von Karstedt
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Anne K Voss
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | | | - Domagoj Vucic
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, CA, USA
| | - Daniela Vuri
- Department of Experimental Medicine, University of Rome Tor Vergata, TOR, Rome, Italy
| | - Erwin F Wagner
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Henning Walczak
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, London, UK
| | - David Wallach
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Ruoning Wang
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Ying Wang
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Achim Weber
- University of Zurich and University Hospital Zurich, Department of Pathology and Molecular Pathology, Zurich, Switzerland
- University of Zurich, Institute of Molecular Cancer Research, Zurich, Switzerland
| | - Will Wood
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Huang-Tian Yang
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Zahra Zakeri
- Queens College and Graduate Center, City University of New York, Flushing, NY, USA
| | - Joanna E Zawacka-Pankau
- Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
- Department of Biochemistry, Laboratory of Biophysics and p53 protein biology, Medical University of Warsaw, Warsaw, Poland
| | - Lin Zhang
- Department of Pharmacology & Chemical Biology, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Haibing Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Boris Zhivotovsky
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Wenzhao Zhou
- Laboratory of Cell Engineering, Institute of Biotechnology, Beijing, China
- Research Unit of Cell Death Mechanism, 2021RU008, Chinese Academy of Medical Science, Beijing, China
| | - Mauro Piacentini
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
- National Institute for Infectious Diseases IRCCS "Lazzaro Spallanzani", Rome, Italy
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
10
|
Shoshan-Barmatz V, Arif T, Shteinfer-Kuzmine A. Apoptotic proteins with non-apoptotic activity: expression and function in cancer. Apoptosis 2023; 28:730-753. [PMID: 37014578 PMCID: PMC10071271 DOI: 10.1007/s10495-023-01835-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2023] [Indexed: 04/05/2023]
Abstract
Apoptosis is a process of programmed cell death in which a cell commits suicide while maintaining the integrity and architecture of the tissue as a whole. Apoptosis involves activation of one of two major pathways: the extrinsic pathway, where extracellular pro-apoptotic signals, transduced through plasma membrane death receptors, activate a caspase cascade leading to apoptosis. The second, the intrinsic apoptotic pathway, where damaged DNA, oxidative stress, or chemicals, induce the release of pro-apoptotic proteins from the mitochondria, leading to the activation of caspase-dependent and independent apoptosis. However, it has recently become apparent that proteins involved in apoptosis also exhibit non-cell death-related physiological functions that are related to the cell cycle, differentiation, metabolism, inflammation or immunity. Such non-conventional activities were predominantly reported in non-cancer cells although, recently, such a dual function for pro-apoptotic proteins has also been reported in cancers where they are overexpressed. Interestingly, some apoptotic proteins translocate to the nucleus in order to perform a non-apoptotic function. In this review, we summarize the unconventional roles of the apoptotic proteins from a functional perspective, while focusing on two mitochondrial proteins: VDAC1 and SMAC/Diablo. Despite having pro-apoptotic functions, these proteins are overexpressed in cancers and this apparent paradox and the associated pathophysiological implications will be discussed. We will also present possible mechanisms underlying the switch from apoptotic to non-apoptotic activities although a deeper investigation into the process awaits further study.
Collapse
Affiliation(s)
- Varda Shoshan-Barmatz
- Department of Life Sciences, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel.
- National Institute for Biotechnology in the Negev, Beer Sheva, Israel.
| | - Tasleem Arif
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | |
Collapse
|
11
|
Amestoy A, Baudrillard C, Briot K, Pizano A, Bouvard M, Lai MC. Steroid hormone pathways, vitamin D and autism: a systematic review. J Neural Transm (Vienna) 2023; 130:207-241. [PMID: 36752873 DOI: 10.1007/s00702-022-02582-6] [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: 10/13/2022] [Accepted: 12/23/2022] [Indexed: 02/09/2023]
Abstract
The origins of the male preponderance in autism incidence remain unclear. The idea that perinatal factors associated with sex differentiation (e.g., steroid hormone pathways) may increase the possibility of the emergence of autism is complementary to the hypothesis that female individuals are intrinsically less likely to develop autism. Empirical evidence for the mechanistic roles of in utero steroid hormones in autism etiology is accumulating but inconsistent. We conducted a systematic review using rigorous criteria for the measurements of steroids and vitamin D exposure, to summarize the potential contributing roles of prenatal and early postnatal steroids and vitamin D alterations to the emergence of autism. We searched PubMed, PsychInfo, Scopus, and included 22 studies for qualitative synthesis. Among them, six studies examined the association of autism diagnoses in offspring and levels of steroids and precursor steroid hormones in the fetal environment, eight studies examined the associations between autism and maternal and fetal blood vitamin D levels during pregnancy and at birth, and eight studies examined the associations between offspring autism diagnoses and maternal hyperandrogenemia diagnosed before pregnancy. We identified promising and complex results regarding the relations between steroid metabolism and autism. The interpretation of findings was limited by the mostly observational study designs, insufficient investigation of the effects of offspring sex, confounders and their cumulative effects on the development of the child, and unclear impact of the timing of steroids exposure and their effects on fetal neurodevelopment.
Collapse
Affiliation(s)
- Anouck Amestoy
- Aquitaine Institute for Cognitive and Integrative Neuroscience, UMR 5287, University of Bordeaux, CNRS, INCIA, Bordeaux, France.
- Centre Hospitalier Charles-Perrens, Pôle Universitaire de Psychiatrie de L'enfant Et de L'adolescent, Bordeaux Cedex, France.
| | - Claire Baudrillard
- Centre Hospitalier Charles-Perrens, Pôle Universitaire de Psychiatrie de L'enfant Et de L'adolescent, Bordeaux Cedex, France
| | - Kellen Briot
- Centre Hospitalier Charles-Perrens, Pôle Universitaire de Psychiatrie de L'enfant Et de L'adolescent, Bordeaux Cedex, France
| | - Adrien Pizano
- Centre Hospitalier Charles-Perrens, Pôle Universitaire de Psychiatrie de L'enfant Et de L'adolescent, Bordeaux Cedex, France
| | - Manuel Bouvard
- Aquitaine Institute for Cognitive and Integrative Neuroscience, UMR 5287, University of Bordeaux, CNRS, INCIA, Bordeaux, France
- Centre Hospitalier Charles-Perrens, Pôle Universitaire de Psychiatrie de L'enfant Et de L'adolescent, Bordeaux Cedex, France
| | - Meng-Chuan Lai
- The Margaret and Wallace McCain Centre for Child, Youth & Family Mental Health and Azrieli Adult Neurodevelopmental Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Department of Psychiatry and Autism Research Unit, The Hospital for Sick Children, Toronto, Canada
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK
- Department of Psychiatry, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| |
Collapse
|
12
|
Sener EF, Dana H, Tahtasakal R, Hamurcu Z, Guler A, Tufan E, Doganyigit Z, Rassoulzadegan M. Partial changes in apoptotic pathways in hippocampus and hypothalamus of Cc2d1a heterozygous. Metab Brain Dis 2023; 38:531-541. [PMID: 36454503 DOI: 10.1007/s11011-022-01125-y] [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: 08/01/2022] [Accepted: 11/10/2022] [Indexed: 12/02/2022]
Abstract
Alterations in the apoptosis pathway have been linked to changes in serotonin levels seen in autistic patients. Cc2d1a is a repressor of the HTR1A gene involved in the serotonin pathway. The hippocampus and hypothalamus of Cc2d1a ± mice were analyzed for the expression of apoptosis markers (caspase 3, 8 and 9). Gender differences were observed in the expression levels of the three caspases consistent with some altered activity in the open-field assay. The number of apoptotic cells was significantly increased. We concluded that apoptotic pathways are only partially affected in the pathogenesis of the Cc2d1a heterozygous mouse model. A) Apoptosis is suppressed because the cell does not receive a death signal, or the receptor cannot activate the caspase 8 pathway despite the death signal. B) Since Caspase 8 and Caspase 3 expression is downregulated in our mouse model, the mechanism of apoptosis is not activated.
Collapse
Affiliation(s)
- Elif Funda Sener
- Department of Medical Biology, Erciyes University Medical Faculty, 38039, Kayseri, Turkey.
- Erciyes University Genome and Stem Cell Center (GENKOK), Kayseri, Turkey.
| | - Halime Dana
- Department of Medical Biology, Erciyes University Medical Faculty, 38039, Kayseri, Turkey
- Erciyes University Genome and Stem Cell Center (GENKOK), Kayseri, Turkey
| | - Reyhan Tahtasakal
- Department of Medical Biology, Erciyes University Medical Faculty, 38039, Kayseri, Turkey
- Erciyes University Genome and Stem Cell Center (GENKOK), Kayseri, Turkey
| | - Zuhal Hamurcu
- Department of Medical Biology, Erciyes University Medical Faculty, 38039, Kayseri, Turkey
- Erciyes University Genome and Stem Cell Center (GENKOK), Kayseri, Turkey
| | - Ahsen Guler
- Department of Medical Biology, Erciyes University Medical Faculty, 38039, Kayseri, Turkey
- Erciyes University Genome and Stem Cell Center (GENKOK), Kayseri, Turkey
| | - Esra Tufan
- Erciyes University Genome and Stem Cell Center (GENKOK), Kayseri, Turkey
| | - Zuleyha Doganyigit
- Department of Histology and Embryology, Bozok University Medical Faculty, 66100, Yozgat, Turkey
| | - Minoo Rassoulzadegan
- Erciyes University Genome and Stem Cell Center (GENKOK), Kayseri, Turkey
- INSERM-CNRS, IRCAN, Universite Cote d'Azur (UCA), 06107, Nice, France
| |
Collapse
|
13
|
Kim SR, Park Y, Li M, Kim YK, Lee S, Son SY, Lee S, Lee JS, Lee CH, Park HH, Lee JY, Hong S, Cho YC, Kim JW, Yoo HM, Cho N, Lee HS, Lee SH. Anti-inflammatory effect of Ailanthus altissima (Mill.) Swingle leaves in lipopolysaccharide-stimulated astrocytes. JOURNAL OF ETHNOPHARMACOLOGY 2022; 286:114258. [PMID: 34271112 DOI: 10.1016/j.jep.2021.114258] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/18/2021] [Accepted: 05/25/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Activated astrocytes are involved in the progression of neurodegenerative diseases. Traditionally, Ailanthus altissima (Mill.) Swingle, widely distributed in East Asia, has been used as a medicine for the treatment of fever, gastric diseases, and inflammation. Although A. altissima has been reported to play an anti-inflammatory role in peripheral tissues or cells, its role in the central nervous system (CNS) remains unclear. AIM OF THE STUDY In the present study, we investigated the anti-inflammatory effects and mechanism of action of A. altissima in primary astrocytes stimulated by lipopolysaccharide (LPS). MATERIALS AND METHODS A nitrite assay was used to measure nitric oxide (NO) production, and the tetrazolium salt 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay was performed to determine cytotoxicity. The expression levels of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and mitogen-activated protein kinase (MAPK) were determined with western blotting. Reverse-transcription PCR was used to assess the expression of inflammatory cytokines. The levels of reactive oxygen species were measured using 2,7-dichlorodihydrofluorescein diacetate. Luciferase assay and immunocytochemistry were used for assessing nuclear factor-kappa B (NF-κB) transcription and p65 localization, respectively. Memory and social interaction were analyzed using the Y-maze and three-chamber tests, respectively. RESULTS The ethanol extract of A. altissima leaves (AAE) inhibited iNOS and COX-2 expression in LPS-stimulated astrocytes. Moreover, AAE reduced the transcription of various proinflammatory mediators, hindered NF-κB activation, and suppressed extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) activation without p38 activation. Ultra-high performance liquid chromatography with mass spectrometry analysis revealed that AAE comprised ethyl gallate, quercetin, and kaempferol, along with luteolin, which has anti-inflammatory properties, and repressed LPS-induced nitrite levels and the nuclear translocation of p65. Finally, oral administration of AAE attenuated LPS-induced memory and social impairment in mice and repressed LPS-induced ERK and JNK activation in the cortices of mice. CONCLUSION AAE could have therapeutic uses in the treatment of neuroinflammatory diseases via suppression of astrocyte activation.
Collapse
Affiliation(s)
- Sung Rae Kim
- Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Yongun Park
- Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Mo Li
- Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Yeong Kyeong Kim
- Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Sunmin Lee
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Su Young Son
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Sarah Lee
- National Institute of Biological Resources, Environmental Research Complex, 42 Hwangyeong-ro, Seo-gu, Incheon, 22755, Republic of Korea
| | - Jong Seok Lee
- National Institute of Biological Resources, Environmental Research Complex, 42 Hwangyeong-ro, Seo-gu, Incheon, 22755, Republic of Korea
| | - Choong Hwan Lee
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Hyun Ho Park
- Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Ji-Yun Lee
- Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Sungguan Hong
- Department of Chemistry, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Young-Chang Cho
- College of Pharmacy, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Jung-Woong Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Hee Min Yoo
- Center for Bioanalysis, Korea Research Institute of Standards and Science, Daejeon, 34113, Republic of Korea
| | - Namki Cho
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyun-Shik Lee
- KNU-Center for Nonlinear Dynamics, CMRI, BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Sung Hoon Lee
- Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea.
| |
Collapse
|
14
|
El Hajj A, Herzine A, Calcagno G, Désor F, Djelti F, Bombail V, Denis I, Oster T, Malaplate C, Vigier M, Kaminski S, Pauron L, Corbier C, Yen FT, Lanhers MC, Claudepierre T. Targeted Suppression of Lipoprotein Receptor LSR in Astrocytes Leads to Olfactory and Memory Deficits in Mice. Int J Mol Sci 2022; 23:ijms23042049. [PMID: 35216163 PMCID: PMC8878779 DOI: 10.3390/ijms23042049] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/05/2022] [Accepted: 02/11/2022] [Indexed: 12/25/2022] Open
Abstract
Perturbations of cholesterol metabolism have been linked to neurodegenerative diseases. Glia–neuron crosstalk is essential to achieve a tight regulation of brain cholesterol trafficking. Adequate cholesterol supply from glia via apolipoprotein E-containing lipoproteins ensures neuronal development and function. The lipolysis-stimulated lipoprotein receptor (LSR), plays an important role in brain cholesterol homeostasis. Aged heterozygote Lsr+/− mice show altered brain cholesterol distribution and increased susceptibility to amyloid stress. Since LSR expression is higher in astroglia as compared to neurons, we sought to determine if astroglial LSR deficiency could lead to cognitive defects similar to those of Alzheimer’s disease (AD). Cre recombinase was activated in adult Glast-CreERT/lsrfl/fl mice by tamoxifen to induce astroglial Lsr deletion. Behavioral phenotyping of young and old astroglial Lsr KO animals revealed hyperactivity during the nocturnal period, deficits in olfactory function affecting social memory and causing possible apathy, as well as visual memory and short-term working memory problems, and deficits similar to those reported in neurodegenerative diseases, such as AD. Furthermore, GFAP staining revealed astroglial activation in the olfactory bulb. Therefore, astroglial LSR is important for working, spatial, and social memory related to sensory input, and represents a novel pathway for the study of brain aging and neurodegeneration.
Collapse
Affiliation(s)
- Aseel El Hajj
- UR AFPA Laboratory, Qualivie Team, University of Lorraine, 54505 Vandoeuvre-lès-Nancy, France; (A.H.); (F.D.); (F.D.); (T.O.); (C.M.); (M.V.); (L.P.); (C.C.); (F.T.Y.); (M.-C.L.)
- Correspondence: (A.E.H.); (T.C.); Tel.: +33-(0)4-8110-6500 (A.E.H.); +33-(0)3-7274-4152 (T.C.)
| | - Ameziane Herzine
- UR AFPA Laboratory, Qualivie Team, University of Lorraine, 54505 Vandoeuvre-lès-Nancy, France; (A.H.); (F.D.); (F.D.); (T.O.); (C.M.); (M.V.); (L.P.); (C.C.); (F.T.Y.); (M.-C.L.)
| | - Gaetano Calcagno
- UR 7300, Stress Immunity Pathogens Laboratory, Faculty of Medicine, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France; (G.C.); (S.K.)
| | - Frédéric Désor
- UR AFPA Laboratory, Qualivie Team, University of Lorraine, 54505 Vandoeuvre-lès-Nancy, France; (A.H.); (F.D.); (F.D.); (T.O.); (C.M.); (M.V.); (L.P.); (C.C.); (F.T.Y.); (M.-C.L.)
| | - Fathia Djelti
- UR AFPA Laboratory, Qualivie Team, University of Lorraine, 54505 Vandoeuvre-lès-Nancy, France; (A.H.); (F.D.); (F.D.); (T.O.); (C.M.); (M.V.); (L.P.); (C.C.); (F.T.Y.); (M.-C.L.)
| | - Vincent Bombail
- UMR 914, Physiology of Nutrition and Feeding Behaviour, INRAE-Agroparistech-Université Paris-Saclay, 78352 Jouy-en-Josas, France; (V.B.); (I.D.)
| | - Isabelle Denis
- UMR 914, Physiology of Nutrition and Feeding Behaviour, INRAE-Agroparistech-Université Paris-Saclay, 78352 Jouy-en-Josas, France; (V.B.); (I.D.)
| | - Thierry Oster
- UR AFPA Laboratory, Qualivie Team, University of Lorraine, 54505 Vandoeuvre-lès-Nancy, France; (A.H.); (F.D.); (F.D.); (T.O.); (C.M.); (M.V.); (L.P.); (C.C.); (F.T.Y.); (M.-C.L.)
| | - Catherine Malaplate
- UR AFPA Laboratory, Qualivie Team, University of Lorraine, 54505 Vandoeuvre-lès-Nancy, France; (A.H.); (F.D.); (F.D.); (T.O.); (C.M.); (M.V.); (L.P.); (C.C.); (F.T.Y.); (M.-C.L.)
| | - Maxime Vigier
- UR AFPA Laboratory, Qualivie Team, University of Lorraine, 54505 Vandoeuvre-lès-Nancy, France; (A.H.); (F.D.); (F.D.); (T.O.); (C.M.); (M.V.); (L.P.); (C.C.); (F.T.Y.); (M.-C.L.)
| | - Sandra Kaminski
- UR 7300, Stress Immunity Pathogens Laboratory, Faculty of Medicine, University of Lorraine, 54500 Vandœuvre-lès-Nancy, France; (G.C.); (S.K.)
| | - Lynn Pauron
- UR AFPA Laboratory, Qualivie Team, University of Lorraine, 54505 Vandoeuvre-lès-Nancy, France; (A.H.); (F.D.); (F.D.); (T.O.); (C.M.); (M.V.); (L.P.); (C.C.); (F.T.Y.); (M.-C.L.)
| | - Catherine Corbier
- UR AFPA Laboratory, Qualivie Team, University of Lorraine, 54505 Vandoeuvre-lès-Nancy, France; (A.H.); (F.D.); (F.D.); (T.O.); (C.M.); (M.V.); (L.P.); (C.C.); (F.T.Y.); (M.-C.L.)
| | - Frances T. Yen
- UR AFPA Laboratory, Qualivie Team, University of Lorraine, 54505 Vandoeuvre-lès-Nancy, France; (A.H.); (F.D.); (F.D.); (T.O.); (C.M.); (M.V.); (L.P.); (C.C.); (F.T.Y.); (M.-C.L.)
| | - Marie-Claire Lanhers
- UR AFPA Laboratory, Qualivie Team, University of Lorraine, 54505 Vandoeuvre-lès-Nancy, France; (A.H.); (F.D.); (F.D.); (T.O.); (C.M.); (M.V.); (L.P.); (C.C.); (F.T.Y.); (M.-C.L.)
| | - Thomas Claudepierre
- UR AFPA Laboratory, Qualivie Team, University of Lorraine, 54505 Vandoeuvre-lès-Nancy, France; (A.H.); (F.D.); (F.D.); (T.O.); (C.M.); (M.V.); (L.P.); (C.C.); (F.T.Y.); (M.-C.L.)
- Correspondence: (A.E.H.); (T.C.); Tel.: +33-(0)4-8110-6500 (A.E.H.); +33-(0)3-7274-4152 (T.C.)
| |
Collapse
|
15
|
Abujamel TS, Al-Otaibi NM, Abuaish S, AlHarbi RH, Assas MB, Alzahrani SA, Alotaibi SM, El-Ansary A, Aabed K. Different Alterations in Gut Microbiota between Bifidobacterium longum and Fecal Microbiota Transplantation Treatments in Propionic Acid Rat Model of Autism. Nutrients 2022; 14:nu14030608. [PMID: 35276971 PMCID: PMC8838423 DOI: 10.3390/nu14030608] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 12/18/2022] Open
Abstract
Autism spectrum disorders (ASD) consist of a range of neurodevelopmental conditions accompanied by dysbiosis of gut microbiota. Therefore, a number of microbiota manipulation strategies were developed to restore their balance. However, a comprehensive comparison of the various methods on gut microbiota is still lacking. Here, we evaluated the effect of Bifidobacterium (BF) treatment and fecal microbiota transplantation (FT) on gut microbiota in a propionic acid (PPA) rat model of autism using 16S rRNA sequencing. Following PPA treatment, gut microbiota showed depletion of Bacteroidia and Akkermansia accompanied by a concomitant increase of Streptococcus, Lachnospiraceae, and Paraeggerthella. The dysbiosis was predicted to cause increased levels of porphyrin metabolism and impairments of acyl-CoA thioesterase and ubiquinone biosynthesis. On the contrary, BF and FT treatments resulted in a distinct increase of Clostridium, Bifidobacterium, Marvinbryantia, Butyricicoccus, and Dorea. The taxa in BF group positively correlated with vitamin B12 and flagella biosynthesis, while FT mainly enriched flagella biosynthesis. In contrast, BF and FT treatments negatively correlated with succinate biosynthesis, pyruvate metabolism, nitrogen metabolism, beta-Lactam resistance, and peptidoglycan biosynthesis. Therefore, the present study demonstrated that BF and FT treatments restored the PPA-induced dysbiosis in a treatment-specific manner.
Collapse
Affiliation(s)
- Turki S. Abujamel
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Correspondence: ; Tel.: +966-504-545-472
| | - Norah M. Al-Otaibi
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia; (N.M.A.-O.); (S.A.A.); (S.M.A.); (K.A.)
| | - Sameera Abuaish
- Department of Basic Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia;
| | - Rahaf H. AlHarbi
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Mushref B. Assas
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Saleha Ahmad Alzahrani
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia; (N.M.A.-O.); (S.A.A.); (S.M.A.); (K.A.)
| | - Sohailah Masoud Alotaibi
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia; (N.M.A.-O.); (S.A.A.); (S.M.A.); (K.A.)
| | - Afaf El-Ansary
- Central Laboratory, Female Center for Medical Studies and Scientific Section, King Saud University, P.O. Box 22452, Riyadh 11472, Saudi Arabia;
| | - Kawther Aabed
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia; (N.M.A.-O.); (S.A.A.); (S.M.A.); (K.A.)
| |
Collapse
|
16
|
Abuaish S, Al-Otaibi NM, Aabed K, Abujamel TS, Alzahrani SA, Alotaibi SM, Bhat RS, Arzoo S, Algahtani N, Moubayed NM, El-Ansary A. The Efficacy of Fecal Transplantation and Bifidobacterium Supplementation in Ameliorating Propionic Acid-Induced Behavioral and Biochemical Autistic Features in Juvenile Male Rats. J Mol Neurosci 2022; 72:372-381. [PMID: 35094316 DOI: 10.1007/s12031-021-01959-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 12/16/2021] [Indexed: 02/06/2023]
Abstract
Gut microbiota plays a major role in neurological disorders, including autism. Modulation of the gut microbiota through fecal microbiota transplantation (FMT) or probiotic administration, such as Bifidobacteria, is suggested to alleviate autistic symptoms; however, their effects on the brain are not fully examined. We tested both approaches in a propionic acid (PPA) rodent model of autism as treatment strategies. Autism was induced in Sprague-Dawley rats by administering PPA orally (250 mg/kg) for 3 days. Animals were later treated with either saline, FMT, or Bifidobacteria for 22 days. Control animals were treated with saline throughout the study. Social behavior and selected brain biochemical markers related to stress hormones, inflammation, and oxidative stress were assessed. PPA treatment induced social impairments, which was rescued by the treatments. In the brain, Bifidobacteria treatment increased oxytocin relative to control and PPA groups. Moreover, Bifidobacteria treatment rescued the PPA-induced increase in IFN-γ levels. Both treatments increased GST levels, which was diminished by the PPA treatment. These findings indicate the potential of gut microbiota-targeted therapeutics in ameliorating behavioral deficit and underlying neural biochemistry.
Collapse
Affiliation(s)
- Sameera Abuaish
- Department of Basic Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Norah M Al-Otaibi
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Kawther Aabed
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Turki S Abujamel
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Saleha Ahmad Alzahrani
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Sohailah Masoud Alotaibi
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Ramesa Shafi Bhat
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Shaista Arzoo
- Department of Food Science and Nutrition, King Saud University, Riyadh, Saudi Arabia
| | - Norah Algahtani
- Central Research Laboratory, King Saud University Female Campus, P O Box 22452, Prince Turki Road, Riyadh, 22452, Saudi Arabia
| | - Nadine Ms Moubayed
- Botany and Microbiology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Central Research Laboratory, King Saud University Female Campus, P O Box 22452, Prince Turki Road, Riyadh, 22452, Saudi Arabia.
| |
Collapse
|
17
|
Han HJ, Lee J, Lim G, Park J, Gautam R, Jo J, Kim C, Heo Y. Metal arsenic mediated enhancement of type-2 immunity in brains with altered locomotive activities in mice with autism-like behavioral characteristics. Toxicol Res 2022; 38:27-33. [PMID: 35070938 PMCID: PMC8748561 DOI: 10.1007/s43188-021-00104-2] [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: 05/25/2021] [Revised: 07/24/2021] [Accepted: 08/25/2021] [Indexed: 01/03/2023] Open
Abstract
Exposure to metal arsenic (As) has been proposed as a risk factor for autism spectrum disorders (ASDs), which are neurodevelopmental disorders with worldwide increasing in its incidence. In the present study, BTBR T + tf/J (BTBR) mice with ASD-like behavioral characteristics and control highly social FVB mice were orally exposed to 0.1 mM arsenic(III)oxide for 4 weeks, and were compared to investigate neuroimmunological or behavioral abnormalities. IgG1:IgG2a ratios in brain tissues from BTBR mice exposed to As (BTBR-As) were significantly higher than those of BTBR-control mice (BTBR-C), but this change did not occur in FVB mice exposed to As. Levels of IL-4, IFN-γ, IL-1β, IL-17, and TNF-α in brain tissue were lowered in BTBR-As relative to BTBR-C, but this tendency was not observed with FVB mice. BTBR-As mice demonstrated decrease in relative travel distance and time spent in the center vs. the periphery of open field arena compared to BTBR-C. Sociability evaluation using three-way chamber test did not clearly demonstrate As-mediated alteration in social interaction in BTBR mice. These findings suggest the potential for As-driven predominant TH2-like reactivity profile in the brain microenvironment of BTBR mice and for As-mediated locomotive impairment probably associated with ASD.
Collapse
Affiliation(s)
- Ha-Jung Han
- College of Bio and Medical Sciences, Department of Occupational Health, Daegu Catholic University, 13-13 Hayang-ro, Gyeongsan-si, Gyeongbuk Province 38430 South Korea
| | - JaeHee Lee
- College of Bio and Medical Sciences, Department of Occupational Health, Daegu Catholic University, 13-13 Hayang-ro, Gyeongsan-si, Gyeongbuk Province 38430 South Korea
| | - GyeongDong Lim
- College of Bio and Medical Sciences, Department of Occupational Health, Daegu Catholic University, 13-13 Hayang-ro, Gyeongsan-si, Gyeongbuk Province 38430 South Korea
| | - JungEun Park
- College of Bio and Medical Sciences, Department of Occupational Health, Daegu Catholic University, 13-13 Hayang-ro, Gyeongsan-si, Gyeongbuk Province 38430 South Korea
| | - Ravi Gautam
- College of Bio and Medical Sciences, Department of Occupational Health, Daegu Catholic University, 13-13 Hayang-ro, Gyeongsan-si, Gyeongbuk Province 38430 South Korea
| | - JiHun Jo
- College of Bio and Medical Sciences, Department of Occupational Health, Daegu Catholic University, 13-13 Hayang-ro, Gyeongsan-si, Gyeongbuk Province 38430 South Korea
| | - ChangYul Kim
- Graduate School Department of Toxicology, Daegu Catholic University, Gyeongsan, South Korea
| | - Yong Heo
- College of Bio and Medical Sciences, Department of Occupational Health, Daegu Catholic University, 13-13 Hayang-ro, Gyeongsan-si, Gyeongbuk Province 38430 South Korea
- Graduate School Department of Toxicology, Daegu Catholic University, Gyeongsan, South Korea
| |
Collapse
|
18
|
Fourman S, Buesing D, Girvin S, Nashawi H, Ulrich-Lai YM. Limited cheese intake reduces HPA axis and behavioral stress responses in male rats. Physiol Behav 2021; 242:113614. [PMID: 34600921 PMCID: PMC8768985 DOI: 10.1016/j.physbeh.2021.113614] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 10/20/2022]
Abstract
Eating palatable foods reduces behavioral and hypothalamic-pituitary-adrenocortical (HPA) axis responses to stress - an idea referred to by the colloquial term "comfort" food. To study the underlying stress-relieving mechanisms of palatable foods, we previously developed a paradigm of limited sucrose feeding in which male rats are given twice-daily access to a small amount of sucrose drink and subsequently have reduced stress responses. Prior research in humans and rodents implicates high dietary sugars/carbohydrates with reduced stress responsivity. However, it is not clear whether the stress-relieving effects of the limited sucrose paradigm depend upon its macronutrient content. To test this idea, the current work measures stress responses in male rats following the limited intermittent intake of cheese - a highly palatable food that is low in sugar and other carbohydrates. The data show that a history of limited cheese intake (LCI) reduced HPA axis responses to acute psychological (restraint) and physiological (hypoxia) stressors. LCI also reduced behavioral struggling during restraint, increased sociability during a social interaction test, and increased open arm activity in the elevated plus-maze test. Z-score analyses evaluated the extent to which these behavioral effects extended within and across assays, and indicated that there was an overall reduction in stress-related behaviors following LCI. Finally, LCI increased immunolabeling for FosB/deltaFosB (a protein associated with repeated or chronic neuronal activation) in the nucleus accumbens. These results indicate that palatable foods can provide stress blunting regardless of their sugar/carbohydrate composition, and support the idea that food reward per se contributes to stress relief.
Collapse
Affiliation(s)
- Sarah Fourman
- Department of Psychiatry and Behavioral Neuroscience, College of Medicine, University of Cincinnati, Cincinnati OH, 45237, USA
| | - Dana Buesing
- Department of Psychiatry and Behavioral Neuroscience, College of Medicine, University of Cincinnati, Cincinnati OH, 45237, USA; Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati OH, 45237, USA
| | - Sean Girvin
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati OH, 45237, USA
| | - Houda Nashawi
- Neuroscience Graduate Program, College of Medicine, University of Cincinnati, Cincinnati OH, 45237, USA
| | - Yvonne M Ulrich-Lai
- Department of Psychiatry and Behavioral Neuroscience, College of Medicine, University of Cincinnati, Cincinnati OH, 45237, USA; Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati OH, 45237, USA.
| |
Collapse
|
19
|
Baranov SV, Jauhari A, Carlisle DL, Friedlander RM. Two hit mitochondrial-driven model of synapse loss in neurodegeneration. Neurobiol Dis 2021; 158:105451. [PMID: 34298088 DOI: 10.1016/j.nbd.2021.105451] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/21/2021] [Accepted: 07/18/2021] [Indexed: 01/11/2023] Open
Abstract
In healthy neurons, a mitochondrial membrane potential gradient exists whereby membrane potential is highest in the soma and decreases with distance from the nucleus. Correspondingly, distal mitochondria have more oxidative damage and slower protein import than somal mitochondria. Due to these differences, distal mitochondria have an intrinsic first stressor that somal mitochondria do not have, resulting in synaptic mitochondrial vulnerability. A second stressor may result from mutant protein expression, situational stress, or aging, exacerbating vulnerable mitochondria activating stress responses. Under these conditions, distal mitochondria release cytochrome c and mitochondrial DNA, leading to compartmentalized sub-lethal caspase-3 activation and cytokine production. In this two-hit mitochondrial-driven synaptic loss model, synapse vulnerability during neurodegeneration is explained as a superposition of pre-existing lower synaptic mitochondrial membrane potential (hit one) with additional mitochondrial stress (hit two). This two-hit mechanism occurs in synaptic mitochondria, activating signaling pathways leading to synaptic degeneration, as a potential preamble to neuronal death.
Collapse
Affiliation(s)
- Sergei V Baranov
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, United States of America
| | - Abhishek Jauhari
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, United States of America
| | - Diane L Carlisle
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, United States of America
| | - Robert M Friedlander
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, United States of America.
| |
Collapse
|
20
|
Fecal Transplant and Bifidobacterium Treatments Modulate Gut Clostridium Bacteria and Rescue Social Impairment and Hippocampal BDNF Expression in a Rodent Model of Autism. Brain Sci 2021; 11:brainsci11081038. [PMID: 34439657 PMCID: PMC8391663 DOI: 10.3390/brainsci11081038] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 12/18/2022] Open
Abstract
Autism is associated with gastrointestinal dysfunction and gut microbiota dysbiosis, including an overall increase in Clostridium. Modulation of the gut microbiota is suggested to improve autistic symptoms. In this study, we explored the implementation of two different interventions that target the microbiota in a rodent model of autism and their effects on social behavior: the levels of different fecal Clostridium spp., and hippocampal transcript levels. Autism was induced in young Sprague Dawley male rats using oral gavage of propionic acid (PPA) for three days, while controls received saline. PPA-treated animals were divided to receive either saline, fecal transplant from healthy donor rats, or Bifidobacterium for 22 days, while controls continued to receive saline. We found that PPA attenuated social interaction in animals, which was rescued by the two interventions. PPA-treated animals had a significantly increased abundance of fecal C. perfringens with a concomitant decrease in Clostridium cluster IV, and exhibited high hippocampal Bdnf expression compared to controls. Fecal microbiota transplantation or Bifidobacterium treatment restored the balance of fecal Clostridium spp. and normalized the level of Bdnf expression. These findings highlight the involvement of the gut-brain axis in the etiology of autism and propose possible interventions in a preclinical model of autism.
Collapse
|
21
|
Abuaish S, Al-Otaibi NM, Aabed K, Abujamel TS, Alzahrani SA, Alotaibi SM, Bhat RS, Arzoo S, El-Ansary A. The role of sex-differentiated variations in stress hormones, antioxidants, and neuroimmune responses in relation to social interaction impairment in a rodent model of autism. Metab Brain Dis 2021; 36:1369-1379. [PMID: 33864573 DOI: 10.1007/s11011-021-00732-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 04/05/2021] [Indexed: 01/30/2023]
Abstract
Males are more likely to develop autism as a neurodevelopmental disorder than females, but the mechanisms underlying male susceptibility are not fully understood. In this paper, we used a well-characterized propionic acid (PPA) rodent model of autism to study sex differences in stress hormones, antioxidants' status, and the neuroimmune response that may contribute to the preponderance of autism in males. Sprague Dawley rats of both sexes were divided into a saline-treated group as controls and PPA-treated groups, receiving 250 mg/kg of PPA per day for three days. Animals' social behavior was examined using the three-chamber social test. Hormones (ACTH, corticosterone, melatonin, and oxytocin), oxidative stress biomarkers (glutathione, glutathione-S-transferase, and ascorbic acid), and cytokines (IL-6, IL-1α, IL-10, and IFNγ) were measured in the brain tissue of all the animals. The results showed a sex dimorphic social response to PPA treatment, where males were more susceptible to the PPA treatment and exhibited a significant reduction in social behavior with no effects observed in females. Also, sex differences were observed in the levels of hormones, antioxidants, and cytokines. Female rats showed significantly higher corticosterone and lower oxytocin, antioxidants, and cytokine levels than males. The PPA treatment later modulated these baseline differences. Our study indicates that the behavioral manifestation of autism in PPA-treated males and not females could be linked to neural biochemical differences between the sexes at baseline, which might play a protective role in females. Our results can contribute to early intervention strategies and treatments used to control autism, an increasingly prevalent disorder.
Collapse
Affiliation(s)
- Sameera Abuaish
- Department of Basic Sciences, College of Medicine, Princess Nourah bint Abdurahman University, Riyadh, Saudi Arabia
| | - Norah M Al-Otaibi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Kawther Aabed
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Turki S Abujamel
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Saleha Ahmad Alzahrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Sohailah Masoud Alotaibi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Ramesa Shafi Bhat
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Shaista Arzoo
- Department of Food Science and Nutrition, King Saud University, Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Central Laboratory, Female Center for Medical Studies and Scientific Section, King Saud University, P O Box 22452, Riyadh, 11495, Saudi Arabia.
| |
Collapse
|
22
|
Saitoh BY, Tanaka E, Yamamoto N, Kruining DV, Iinuma K, Nakamuta Y, Yamaguchi H, Yamasaki R, Matsumoto K, Kira JI. Early postnatal allergic airway inflammation induces dystrophic microglia leading to excitatory postsynaptic surplus and autism-like behavior. Brain Behav Immun 2021; 95:362-380. [PMID: 33862170 DOI: 10.1016/j.bbi.2021.04.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 04/01/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia play key roles in synaptic pruning, which primarily occurs from the postnatal period to adolescence. Synaptic pruning is essential for normal brain development and its impairment is implicated in neuropsychiatric developmental diseases such as autism spectrum disorders (ASD). Recent epidemiological surveys reported a strong link between ASD and atopic/allergic diseases. However, few studies have experimentally investigated the relationship between allergy and ASD-like manifestations, particularly in the early postnatal period, when allergic disorders occur frequently. Therefore, we aimed to characterize how allergic inflammation in the early postnatal period influences microglia and behavior using mouse models of short- and long-term airway allergy. Male mice were immunized by an intraperitoneal injection of aluminum hydroxide and ovalbumin (OVA) or phosphate-buffered saline (control) on postnatal days (P) 3, 7, and 11, followed by intranasal challenge with OVA or phosphate-buffered saline solution twice a week until P30 or P70. In the hippocampus, Iba-1-positive areas, the size of Iba-1-positive microglial cell bodies, and the ramification index of microglia by Sholl analysis were significantly smaller in the OVA group than in the control group on P30 and P70, although Iba-1-positive microglia numbers did not differ significantly between the two groups. In Iba-1-positive cells, postsynaptic density protein 95 (PSD95)-occupied areas and CD68-occupied areas were significantly decreased on P30 and P70, respectively, in the OVA group compared with the control group. Immunoblotting using hippocampal tissues demonstrated that amounts of PSD95, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor 2, and N-methyl-D-aspartate (NMDA) receptor 2B were significantly increased in the OVA group compared with the control group on P70, and a similar increasing trend for PSD95 was observed on P30. Neurogenesis was not significantly different between the two groups on P30 or P70 by doublecortin immunohistochemistry. The social preference index was significantly lower in the three chamber test and the number of buried marbles was significantly higher in the OVA group than in the control group on P70 but not on P30, whereas locomotion and anxiety were not different between the two groups. Compared with the control group, serum basal corticosterone levels were significantly elevated and hippocampal glucocorticoid receptor (GR) amounts and nuclear GR translocation in microglia, but not in neurons or astrocytes, were significantly decreased in the OVA group on P70 but not on P30. Gene set enrichment analysis of isolated microglia revealed that genes related to immune responses including Toll-like receptor signaling and chemokine signaling pathways, senescence, and glucocorticoid signaling were significantly upregulated in the OVA group compared with the control group on P30 and P70. These findings suggest that early postnatal allergic airway inflammation induces dystrophic microglia that exhibit defective synaptic pruning upon short- and long-term allergen exposure. Furthermore, long-term allergen exposure induced excitatory postsynaptic surplus and ASD-like behavior. Hypothalamo-pituitary-adrenal axis activation and the compensatory downregulation of microglial GR during long-term allergic airway inflammation may also facilitate these changes.
Collapse
Affiliation(s)
- Ban-Yu Saitoh
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eizo Tanaka
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Norio Yamamoto
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daan van Kruining
- School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, Netherlands
| | - Kyoko Iinuma
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuko Nakamuta
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroo Yamaguchi
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichiro Matsumoto
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Translational Neuroscience Center, Graduate School of Medicine, and School of Pharmacy at Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Ookawa, Fukuoka 831-8501, Japan; Department of Neurology, Brain and Nerve Center, Fukuoka Central Hospital, International University of Health and Welfare, 2-6-11 Yakuin, Chuo-ku, Fukuoka 810-0022, Japan.
| |
Collapse
|
23
|
Xu J, Marshall JJ, Kraniotis S, Nomura T, Zhu Y, Contractor A. Genetic disruption of Grm5 causes complex alterations in motor activity, anxiety and social behaviors. Behav Brain Res 2021; 411:113378. [PMID: 34029630 DOI: 10.1016/j.bbr.2021.113378] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/11/2021] [Accepted: 05/19/2021] [Indexed: 12/14/2022]
Abstract
Autism is a neurodevelopmental disorder characterized by impaired social interactions and restricted and repetitive behaviors. Although group 1 metabotropic glutamate receptors (mGluRs), and in particular mGluR5, have been extensively proposed as potential targets for intervention in autism and other neurodevelopmental disorders, there has not been a comprehensive analysis of the effect of mGluR5 loss on behaviors typically assessed in autism mouse models thought to be correlates of behavioral symptoms of human disorders. Here we present a behavioral characterization of mice with complete or partial loss of mGluR5 (homozygous or heterozygous null mutations in Grm5 gene). We tested several autism related behaviors including social interaction, repetitive grooming, digging and locomotor behaviors. We found that digging and marble burying behaviors were almost completely abolished in mGluR5 ko mice, although self-grooming was not altered. Social interaction was impaired in ko but not in heterozygote (het) mice. In tests of locomotor activity and anxiety related behaviors, mGluR5 ko mice exhibited hyperactivity and reduced anxiety in the open field test but unexpectedly, showed hypoactivity in the elevated zero-maze test. There was no impairment in motor learning in the accelerating rotarod in both ko and het mutant. Together these results provide support for the importance of mGluR5 in motor and social behaviors that are specifically affected in autism disorders.
Collapse
Affiliation(s)
- Jian Xu
- Department of Physiology, Northwestern University Feinberg School of Medicine, United States.
| | - John J Marshall
- Department of Physiology, Northwestern University Feinberg School of Medicine, United States
| | - Stephen Kraniotis
- Department of Physiology, Northwestern University Feinberg School of Medicine, United States
| | - Toshihiro Nomura
- Department of Physiology, Northwestern University Feinberg School of Medicine, United States
| | - Yongling Zhu
- Department of Physiology, Northwestern University Feinberg School of Medicine, United States
| | - Anis Contractor
- Department of Physiology, Northwestern University Feinberg School of Medicine, United States; Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Chicago, IL, 60611, United States.
| |
Collapse
|
24
|
Adu-Nti F, Gao X, Wu JM, Li J, Iqbal J, Ahmad R, Ma XM. Osthole Ameliorates Estrogen Deficiency-Induced Cognitive Impairment in Female Mice. Front Pharmacol 2021; 12:641909. [PMID: 34025413 PMCID: PMC8134730 DOI: 10.3389/fphar.2021.641909] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/04/2021] [Indexed: 12/24/2022] Open
Abstract
Loss of endogenous estrogen and dysregulation of the estrogen receptor signaling pathways are associated with an increase in risk for cognitive deficit and depression in women after menopause. Estrogen therapy for menopause increases the risk of breast and ovarian cancers, and stroke. Therefore, it is critical to find an alternate treatment for menopausal women. Osthole (OST), a coumarin, has been reported to have neuroprotective effects. This study examined whether OST improves ovariectomy (OVX)-induced cognitive impairment, and alleviates anxiety- and depression-like behaviors induced by OVX in mice. Adult female C57BL/6J mice were ovariectomized and then treated with OST at a dose of 30 mg/kg for 14 days. At the end of the treatment period, behavioral tests were used to evaluate spatial learning and memory, recognition memory, anxiety- and depression-like behaviors. A cohort of the mice were sacrificed after 14 days of OST treatment and their hippocampi were collected for measurement of the proteins of interest using western blot. OVX-induced alteration in the levels of proteins was accompanied by cognitive deficit, anxiety- and depression-like behaviors. OST treatment improved cognitive deficit, alleviated anxiety- and depression-like behaviors induced by OVX, and reversed OVX-induced alterations in the levels of synaptic proteins and ERα, BDNF, TrKB, p-CREB, p-Akt and Rac1 in the hippocampus. Therefore, reversal of OVX-induced decrease in the levels of hippocampal proteins by OST might contribute to the effects of OST on improving cognitive deficit and alleviating anxiety- and depression-like behaviors induced by OVX.
Collapse
Affiliation(s)
- Frank Adu-Nti
- Department of Neuroscience, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xu Gao
- Department of Neuroscience, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Jia-Min Wu
- Department of Neuroscience, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Jing Li
- Department of Neuroscience, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Javed Iqbal
- Department of Neuroscience, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Riaz Ahmad
- Department of Neuroscience, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xin-Ming Ma
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States
| |
Collapse
|
25
|
Rymut HE, Bolt CR, Caputo MP, Houser AK, Antonson AM, Zimmerman JD, Villamil MB, Southey BR, Rund LA, Johnson RW, Rodriguez-Zas SL. Long-Lasting Impact of Maternal Immune Activation and Interaction With a Second Immune Challenge on Pig Behavior. Front Vet Sci 2020; 7:561151. [PMID: 33330688 PMCID: PMC7732429 DOI: 10.3389/fvets.2020.561151] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 10/28/2020] [Indexed: 01/30/2023] Open
Abstract
The combined effects on pig behavior of maternal immune challenge during gestation followed by a second immune challenge later in life have not been studied. Porcine reproductive and respiratory syndrome virus (PRRSV) infection during gestation can elicit maternal immune activation (MIA) yet the interactions with the offspring response to a second immune challenge after birth remains unexplored. Knowledge on the response to viral challenges in rodents has been gained through the use of the viral mimetic polyinosinic-polycytidylic acid (Poly(I:C)), yet the effects of this immune stimulant on pig behavior have not been assessed. This study advances the understanding of the combined effect of MIA and a second immune challenge later in life on female and male pig behavior. Three complementary experiments enabled the development of an effective Poly(I:C) challenge in pigs, and testing the interaction between PRRSV-elicited MIA, Poly(I:C) challenge at 60 days of age, and sex on behaviors. Individual-level observations on sickness, locomotor, and social behaviors were measured 1-3 h after Poly(I:C) challenge. Vomiting, panting, lethargy, walking, laying, playing, and touching behaviors were analyzed using generalized linear mixed effect models. Results indicated that a Poly(I:C) dose of 1 mg/kg within 1 h after injection increased the incidence of laying and sickness behavior. The Poly(I:C) challenge decreased the incidence of locomotor behaviors and activity levels. Pigs exposed to MIA had lower rates of social behaviors such as playing. The combined effect of PRRSV-elicited MIA and Poly(I:C) immune challenge further sensitized the pigs to behavior disruption across sexes including changes in sternal and lateral laying, walking, lethargy, and touching incidence. Notably, the effects of Poly(I:C) immune challenge alone on behaviors tended to be more extreme in males, whereas the effects of Poly(I:C) following MIA tended to be more extreme in females. Our findings demonstrate that MIA and Poly(I:C) affected behaviors, and the viral mimetic effects shortly after injection can offer insights into the prolonged effect of postnatal viral infections on feeding, social interactions and health status. Management practices that reduce the likelihood of gestational diseases and accommodate for behavioral disruptions in the offspring can minimize the impact of MIA.
Collapse
Affiliation(s)
- Haley E Rymut
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Courtni R Bolt
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Megan P Caputo
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Alexandra K Houser
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Adrienne M Antonson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Jalisa D Zimmerman
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Maria B Villamil
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Bruce R Southey
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Laurie A Rund
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Rodney W Johnson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Sandra L Rodriguez-Zas
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Department of Crop Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,C. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
26
|
Klibaite U, Shaevitz JW. Paired fruit flies synchronize behavior: Uncovering social interactions in Drosophila melanogaster. PLoS Comput Biol 2020; 16:e1008230. [PMID: 33021989 PMCID: PMC7567355 DOI: 10.1371/journal.pcbi.1008230] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 10/16/2020] [Accepted: 08/09/2020] [Indexed: 11/19/2022] Open
Abstract
Social behaviors are ubiquitous and crucial to an animal's survival and success. The behaviors an animal performs in a social setting are affected by internal factors, inputs from the environment, and interactions with others. To quantify social behaviors, we need to measure both the stochastic nature of the behavior of isolated individuals and how this behavioral repertoire changes as a function of the environment and interactions between individuals. We probed the behavior of male and female fruit flies in a circular arena as individuals and within all possible pairings. By combining measurements of the animals' position in the arena with an unsupervised analysis of their behaviors, we define the effects of position in the environment and the presence of a partner on locomotion, grooming, singing, and other behaviors that make up an animal's repertoire. We find that geometric context tunes behavioral preference, pairs of animals synchronize their behavioral preferences across shared trials, and paired individuals display signatures of behavioral mimicry.
Collapse
Affiliation(s)
- Ugne Klibaite
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey, USA
| | - Joshua W Shaevitz
- Department of Physics, Princeton University, Princeton, New Jersey, USA
| |
Collapse
|
27
|
Carvalho FR, Nóbrega CDR, Martins AT. Mapping gene expression in social anxiety reveals the main brain structures involved in this disorder. Behav Brain Res 2020; 394:112808. [PMID: 32707139 DOI: 10.1016/j.bbr.2020.112808] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/24/2020] [Accepted: 07/10/2020] [Indexed: 12/18/2022]
Abstract
Social Anxiety Disorder (SAD) is characterized by emotional and attentional biases as well as distorted negative self-beliefs. According this, we proposed to identify the brain structures and hub genes involved in SAD. An analysis in Pubmed and TRANSFAC was conducted and 72 genes were identified. Using Microarray data, from Allen Human Brain Atlas, it was possible to identify three modules of co-expressed genes from our gene set (R package WGCNA). Higher mean gene expression was found in cortico-medial group, basomedial nucleus, ATZ in amygdala and in head and tail of the caudate nucleus, nucleus accumbens and putamen in striatum. Our enrichment analysis identified the followed hub genes: DRD2, HTR1A, JUN, SP1 and HDAC4. We suggest that SAD is explained by delayed extinction of circuitry for conditioned fear. Caused by reduced activation of the dopaminergic and serotonergic systems,due diminished expectation of reward during social interactions.
Collapse
Affiliation(s)
- Filipe Ricardo Carvalho
- Department of Biomedical Sciences and Medicine, University of Algarve, Portugal; University of Algarve Campus De Gambelas, 8005-139 Faro, Portugal.
| | - Clévio David Rodrigues Nóbrega
- Center for Biomedicine Research (CBMR), University of Algarve, Portugal; Department of Biomedical Sciences and Medicine, University of Algarve, Portugal; Algarve Biomedical Center (ABC); University of Algarve Campus De Gambelas, 8005-139 Faro, Portugal
| | - Ana Teresa Martins
- Center for Biomedicine Research (CBMR), University of Algarve, Portugal; Department of Psychology and Education Sciences, University of Algarve, Portugal; University of Algarve Campus De Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
28
|
Piccin A, Contarino A. Sex-linked roles of the CRF 1 and the CRF 2 receptor in social behavior. J Neurosci Res 2020; 98:1561-1574. [PMID: 32468598 DOI: 10.1002/jnr.24629] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 03/17/2020] [Accepted: 04/03/2020] [Indexed: 11/08/2022]
Abstract
Dysfunctional social behavior is a major clinical feature of mood, anxiety, autism spectrum, and substance-related disorders, and may dramatically contribute to the poor outcome of these diseases. Nevertheless, the mechanisms underlying social behavior deficits are still largely unknown. The corticotropin-releasing factor (CRF) system, a major coordinator of the stress response, has been hypothesized to modulate social behavior. CRF signaling is mediated by two receptor types, termed CRF1 and CRF2 . Using the three-chamber task for sociability (i.e., preference for an unfamiliar conspecific vs. an object), this study demonstrates that CRF2 receptor null mutation (CRF2 -/-) reduces sociability in female mice but increases it in male mice. Both female and male CRF2 -/- mice display a preference for social odor cues over neutral cues, indicating that sex- and CRF2 receptor-dependent sociability is not due to altered olfaction or impaired social cues discrimination. Moreover, treatment with the CRF1 receptor-preferring antagonist, antalarmin, consistently induces sociability in non-social mice but disrupts it in social mice, independently of CRF2 receptor deficiency. Sex, CRF2 receptor deficiency, or antalarmin affect locomotor activity during the three-chamber test. However, throughout the study CRF1 and CRF2 receptor-linked sociability is independent of locomotor activity. The present findings highlight major functions for the CRF system in the regulation of social behavior. Moreover, they provide initial evidence of sex-linked roles for the CRF1 and the CRF2 receptor, emphasizing the importance of sex as a major biological variable to be taken into consideration in preclinical and clinical studies.
Collapse
Affiliation(s)
- Alessandro Piccin
- INCIA, UMR 5287, Université de Bordeaux, Bordeaux, France.,CNRS, INCIA, UMR 5287, Bordeaux, France
| | - Angelo Contarino
- INCIA, UMR 5287, Université de Bordeaux, Bordeaux, France.,CNRS, INCIA, UMR 5287, Bordeaux, France
| |
Collapse
|
29
|
Kim Y, Noh YW, Kim K, Yang E, Kim H, Kim E. IRSp53 Deletion in Glutamatergic and GABAergic Neurons and in Male and Female Mice Leads to Distinct Electrophysiological and Behavioral Phenotypes. Front Cell Neurosci 2020; 14:23. [PMID: 32116566 PMCID: PMC7026675 DOI: 10.3389/fncel.2020.00023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 01/27/2020] [Indexed: 12/25/2022] Open
Abstract
IRSp53 (also known as BAIAP2) is an abundant excitatory postsynaptic scaffolding protein implicated in autism spectrum disorders (ASD), schizophrenia, and attention-deficit/hyperactivity disorder (ADHD). IRSp53 is expressed in different cell types across different brain regions, although it remains unclear how IRSp53 deletion in different cell types affects brain functions and behaviors in mice. Here, we deleted IRSp53 in excitatory and inhibitory neurons in mice and compared resulting phenotypes in males and females. IRSp53 deletion in excitatory neurons driven by Emx1 leads to strong social deficits and hyperactivity without affecting anxiety-like behavior, whereas IRSp53 deletion in inhibitory neurons driven by Viaat has minimal impacts on these behaviors in male mice. In female mice, excitatory neuronal IRSp53 deletion induces hyperactivity but moderate social deficits. Excitatory neuronal IRSp53 deletion in male mice induces an increased ratio of evoked excitatory and inhibitory synaptic transmission (E/I ratio) in layer V pyramidal neurons in the prelimbic region of the medial prefrontal cortex (mPFC), whereas the same mutation does not alter the E/I ratio in female neurons. These results suggest that IRSp53 deletion in excitatory and inhibitory neurons and in male and female mice has distinct impacts on behaviors and synaptic transmission.
Collapse
Affiliation(s)
- Yangsik Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Young Woo Noh
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Kyungdeok Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Esther Yang
- Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Hyun Kim
- Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea.,Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| |
Collapse
|
30
|
Chu CH, Chen JS, Chuang PC, Su CH, Chan YL, Yang YJ, Chiang YT, Su YY, Gean PW, Sun HS. TIAM2S as a novel regulator for serotonin level enhances brain plasticity and locomotion behavior. FASEB J 2020; 34:3267-3288. [PMID: 31908036 DOI: 10.1096/fj.201901323r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 10/18/2019] [Accepted: 12/23/2019] [Indexed: 11/11/2022]
Abstract
TIAM2S, the short form of human T-cell lymphoma invasion and metastasis 2, can have oncogenic effects when aberrantly expressed in the liver or lungs. However, it is also abundant in healthy, non-neoplastic brain tissue, in which its primary function is still unknown. Here, we examined the neurobiological and behavioral significance of human TIAM2S using the human brain protein panels, a human NT2/D1-derived neuronal cell line model (NT2/N), and transgenic mice that overexpress human TIAM2S (TIAM2S-TG). Our data reveal that TIAM2S exists primarily in neurons of the restricted brain areas around the limbic system and in well-differentiated NT2/N cells. Functional studies revealed that TIAM2S has no guanine nucleotide exchange factor (GEF) activity and is mainly located in the nucleus. Furthermore, whole-transcriptome and enrichment analysis with total RNA sequencing revealed that TIAM2S-knockdown (TIAM2S-KD) was strongly associated with the cellular processes of the brain structural development and differentiation, serotonin-related signaling, and the diseases markers representing neurobehavioral developmental disorders. Moreover, TIAM2S-KD cells display decreased neurite outgrowth and reduced serotonin levels. Moreover, TIAM2S overexpressing TG mice show increased number and length of serotonergic fibers at early postnatal stage, results in higher serotonin levels at both the serum and brain regions, and higher neuroplasticity and hyperlocomotion in latter adulthood. Taken together, our results illustrate the non-oncogenic functions of human TIAM2S and demonstrate that TIAM2S is a novel regulator of serotonin level, brain neuroplasticity, and locomotion behavior.
Collapse
Affiliation(s)
- Chun-Hsien Chu
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jia-Shing Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Chin Chuang
- Department of Medical Research, Chang Gung Memorial Hospital, Kaohsiung, Taiwan.,Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Hao Su
- Institute for Translational Research in Biomedicine, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Ya-Ling Chan
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Ying-Ju Yang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Ting Chiang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Ya Su
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Po-Wu Gean
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan.,Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - H Sunny Sun
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
31
|
Ariyasiri K, Choi TI, Kim OH, Hong TI, Gerlai R, Kim CH. Pharmacological (ethanol) and mutation (sam2 KO) induced impairment of novelty preference in zebrafish quantified using a new three-chamber social choice task. Prog Neuropsychopharmacol Biol Psychiatry 2019; 88:53-65. [PMID: 29958859 DOI: 10.1016/j.pnpbp.2018.06.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/06/2018] [Accepted: 06/19/2018] [Indexed: 01/08/2023]
Abstract
Social behavior is a fundamental aspect of our own species, a feature without which our society would not function. There are numerous human brain disorders associated with abnormal social behavior, among them are the autism spectrum disorders whose causal factors include a genetic component. Environmental factors, including drugs of abuse such as alcohol, also contribute to numerous abnormalities related to social behavior. Several such disorders have been modeled using laboratory animals. Perhaps one of the newest among them is the zebrafish. However, the paucity of standardized behavioral assays specifically developed for the zebrafish have hindered progress. Here, we present a newly developed zebrafish behavioral paradigm, the three-chamber social choice task. This task, which was adapted from a murine model, assesses sociality and social novelty preference in zebrafish in three phases: habituation, phase-I to evaluate sociality, and phase-II to quantify social novelty preference. Test fish are placed in the middle chamber, while conspecifics are introduced to the flanking chambers during phase-I and II. Both male and female zebrafish displayed sociality (preference for conspecifics) during phase-I and social novelty preference (preference for unfamiliar conspecifics) during phase-II. We found the paradigm to be able to detect both environmentally (alcohol) as well as genetically (targeted knock out of sam2) induced alterations of behavioral phenotypes. Although ethanol-treated fish displayed similar levels of sociality to those of control (not alcohol exposed) male and female zebrafish, they were found to exhibit significantly impaired social novelty preference, a finding compatible with altered motivational or perhaps mnemonic processes. Moreover, we found that knock out of sam2, previously shown to lead to emotional dysregulation, also disrupted social novelty preference, while leaving sociality relatively intact. We conclude that our novel behavioral paradigm is appropriate for the modeling and quantification of social behavior deficits in zebrafish.
Collapse
Affiliation(s)
- Krishan Ariyasiri
- Department of Biology, Chungnam National University, Daejeon 34134, South Korea
| | - Tae-Ik Choi
- Department of Biology, Chungnam National University, Daejeon 34134, South Korea
| | - Oc-Hee Kim
- Department of Biology, Chungnam National University, Daejeon 34134, South Korea
| | - Ted Inpyo Hong
- Department of Biology, Chungnam National University, Daejeon 34134, South Korea
| | - Robert Gerlai
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada.
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon 34134, South Korea.
| |
Collapse
|
32
|
Sexually dimorphic behavior, neuronal activity, and gene expression in Chd8-mutant mice. Nat Neurosci 2018; 21:1218-1228. [DOI: 10.1038/s41593-018-0208-z] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 06/21/2018] [Indexed: 12/31/2022]
|
33
|
Narayanan SN, Mohapatra N, John P, K N, Kumar RS, Nayak SB, Bhat PG. Radiofrequency electromagnetic radiation exposure effects on amygdala morphology, place preference behavior and brain caspase-3 activity in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 58:220-229. [PMID: 29413766 DOI: 10.1016/j.etap.2018.01.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/23/2017] [Accepted: 01/23/2018] [Indexed: 06/08/2023]
Abstract
The purpose of the study was to evaluate the changes in amygdala morphology and emotional behaviors, upon exposure to chronic RF-EMR in adolescent rats. Four weeks old male albino Wistar rats were exposed to 900 MHz (power density:146.60 μW/cm2) from a mobile phone in silent-mode for 28 days. Amygdala morphology was studied using cresyl violet, TUNEL and Golgi-Cox staining. Place preference behavior was studied using light/dark chamber test and following this brain caspase-3 activity was determined. Number of healthy neurons was decreased in the basolateral amygdala and cortical amygdala but not in the central amygdala after RF-EMR exposure. It also induced apoptosis in the amygdala. RF-EMR exposure altered dendritic arborization pattern in basolateral amygdala but not in the central amygdala. Altered place preference and hyperactivity-like behavior was evident after RF-EMR exposure, but brain caspase-3 activity did not change. RF-EMR exposure perturbed normal cellular architecture of amygdala and this was associated with altered place preference.
Collapse
Affiliation(s)
- Sareesh Naduvil Narayanan
- Department of Physiology, Melaka Manipal Medical College (Manipal Campus), Manipal University, Manipal, 576104, India.
| | - Nirupam Mohapatra
- Department of Biochemistry, Kasturba Medical College, Manipal University, Manipal, 576104, India
| | - Pamala John
- Department of Biochemistry, Kasturba Medical College, Manipal University, Manipal, 576104, India
| | - Nalini K
- Department of Biochemistry, Kasturba Medical College, Manipal University, Manipal, 576104, India
| | - Raju Suresh Kumar
- Department of Physiology, Melaka Manipal Medical College (Manipal Campus), Manipal University, Manipal, 576104, India
| | - Satheesha B Nayak
- Department of Anatomy, Melaka Manipal Medical College (Manipal Campus), Manipal University, Manipal, 576104, India
| | - P Gopalakrishna Bhat
- Division of Biotechnology, School of Life Sciences, Manipal University, Manipal, 576 104, India
| |
Collapse
|
34
|
Abstract
Brain development diverges in males and females in response to androgen production by the fetal testis. This sexual differentiation of the brain occurs during a sensitive window and induces enduring neuroanatomical and physiological changes that profoundly impact behavior. What we know about the contribution of sex chromosomes is still emerging, highlighting the need to integrate multiple factors into understanding sex differences, including the importance of context. The cellular mechanisms are best modeled in rodents and have provided both unifying principles and surprising specifics. Markedly distinct signaling pathways direct differentiation in specific brain regions, resulting in mosaicism of relative maleness, femaleness, and sameness through-out the brain, while canalization both exaggerates and constrains sex differences. Non-neuronal cells and inflammatory mediators are found in greater number and at higher levels in parts of male brains. This higher baseline of inflammation is speculated to increase male vulnerability to developmental neuropsychiatric disorders that are triggered by inflammation.
Collapse
Affiliation(s)
- Margaret M McCarthy
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
35
|
Tian Y, Yang C, Shang S, Cai Y, Deng X, Zhang J, Shao F, Zhu D, Liu Y, Chen G, Liang J, Sun Q, Qiu Z, Zhang C. Loss of FMRP Impaired Hippocampal Long-Term Plasticity and Spatial Learning in Rats. Front Mol Neurosci 2017; 10:269. [PMID: 28894415 PMCID: PMC5581399 DOI: 10.3389/fnmol.2017.00269] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 08/09/2017] [Indexed: 11/13/2022] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by mutations in the FMR1 gene that inactivate expression of the gene product, the fragile X mental retardation 1 protein (FMRP). In this study, we used clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) technology to generate Fmr1 knockout (KO) rats by disruption of the fourth exon of the Fmr1 gene. Western blotting analysis confirmed that the FMRP was absent from the brains of the Fmr1 KO rats (Fmr1exon4-KO ). Electrophysiological analysis revealed that the theta-burst stimulation (TBS)-induced long-term potentiation (LTP) and the low-frequency stimulus (LFS)-induced long-term depression (LTD) were decreased in the hippocampal Schaffer collateral pathway of the Fmr1exon4-KO rats. Short-term plasticity, measured as the paired-pulse ratio, remained normal in the KO rats. The synaptic strength mediated by the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) was also impaired. Consistent with previous reports, the Fmr1exon4-KO rats demonstrated an enhanced 3,5-dihydroxyphenylglycine (DHPG)-induced LTD in the present study, and this enhancement is insensitive to protein translation. In addition, the Fmr1exon4-KO rats showed deficits in the probe trial in the Morris water maze test. These results demonstrate that deletion of the Fmr1 gene in rats specifically impairs long-term synaptic plasticity and hippocampus-dependent learning in a manner resembling the key symptoms of FXS. Furthermore, the Fmr1exon4-KO rats displayed impaired social interaction and macroorchidism, the results consistent with those observed in patients with FXS. Thus, Fmr1exon4-KO rats constitute a novel rat model of FXS that complements existing mouse models.
Collapse
Affiliation(s)
- Yonglu Tian
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University-IDG/McGovern Institute for Brain Research, Peking UniversityBeijing, China.,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijing, China
| | - Chaojuan Yang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University-IDG/McGovern Institute for Brain Research, Peking UniversityBeijing, China
| | - Shujiang Shang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University-IDG/McGovern Institute for Brain Research, Peking UniversityBeijing, China
| | - Yijun Cai
- CAS Key Laboratory of Primate Neurobiology, Institute of Neuroscience, Chinese Academy of SciencesShanghai, China
| | - Xiaofei Deng
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of SciencesBeijing, China
| | - Jian Zhang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University-IDG/McGovern Institute for Brain Research, Peking UniversityBeijing, China
| | - Feng Shao
- Department of Psychology, Peking UniversityBeijing, China
| | - Desheng Zhu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University-IDG/McGovern Institute for Brain Research, Peking UniversityBeijing, China
| | - Yunbo Liu
- Institute of Laboratory Animal Science, Peking Union Medical College/Chinese Academy of Medical SciencesBeijing, China
| | - Guiquan Chen
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing UniversityNanjing, China
| | - Jing Liang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of SciencesBeijing, China
| | - Qiang Sun
- CAS Key Laboratory of Primate Neurobiology, Institute of Neuroscience, Chinese Academy of SciencesShanghai, China
| | - Zilong Qiu
- CAS Key Laboratory of Primate Neurobiology, Institute of Neuroscience, Chinese Academy of SciencesShanghai, China
| | - Chen Zhang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University-IDG/McGovern Institute for Brain Research, Peking UniversityBeijing, China.,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking UniversityBeijing, China
| |
Collapse
|
36
|
Gris KV, Coutu JP, Gris D. Supervised and Unsupervised Learning Technology in the Study of Rodent Behavior. Front Behav Neurosci 2017; 11:141. [PMID: 28804452 PMCID: PMC5532435 DOI: 10.3389/fnbeh.2017.00141] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/17/2017] [Indexed: 12/17/2022] Open
Abstract
Quantifying behavior is a challenge for scientists studying neuroscience, ethology, psychology, pathology, etc. Until now, behavior was mostly considered as qualitative descriptions of postures or labor intensive counting of bouts of individual movements. Many prominent behavioral scientists conducted studies describing postures of mice and rats, depicting step by step eating, grooming, courting, and other behaviors. Automated video assessment technologies permit scientists to quantify daily behavioral patterns/routines, social interactions, and postural changes in an unbiased manner. Here, we extensively reviewed published research on the topic of the structural blocks of behavior and proposed a structure of behavior based on the latest publications. We discuss the importance of defining a clear structure of behavior to allow professionals to write viable algorithms. We presented a discussion of technologies that are used in automated video assessment of behavior in mice and rats. We considered advantages and limitations of supervised and unsupervised learning. We presented the latest scientific discoveries that were made using automated video assessment. In conclusion, we proposed that the automated quantitative approach to evaluating animal behavior is the future of understanding the effect of brain signaling, pathologies, genetic content, and environment on behavior.
Collapse
Affiliation(s)
- Katsiaryna V Gris
- Gris Lab of Neuroimmunology, Pediatrics, University of SherbrookeSherbrooke, QC, Canada
| | - Jean-Philippe Coutu
- Gris Lab of Neuroimmunology, Pediatrics, University of SherbrookeSherbrooke, QC, Canada
| | - Denis Gris
- Gris Lab of Neuroimmunology, Pediatrics, University of SherbrookeSherbrooke, QC, Canada
| |
Collapse
|
37
|
|
38
|
Garapaty A, Champion JA. Tunable particles alter macrophage uptake based on combinatorial effects of physical properties. Bioeng Transl Med 2017; 2:92-101. [PMID: 29313025 PMCID: PMC5689517 DOI: 10.1002/btm2.10047] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 11/04/2016] [Accepted: 11/07/2016] [Indexed: 12/31/2022] Open
Abstract
The ability to tune phagocytosis of particle-based therapeutics by macrophages can enhance their delivery to macrophages or reduce their phagocytic susceptibility for delivery to non-phagocytic cells. Since phagocytosis is affected by the physical and chemical properties of particles, it is crucial to identify any interplay between physical properties of particles in altering phagocytic interactions. The combinatorial effect of physical properties size, shape and stiffness was investigated on Fc receptor mediated macrophage interactions by fabrication of layer-by-layer tunable particles of constant surface chemistry. Our results highlight how changing particle stiffness affects phagocytic interaction intricately when combined with varying size or shape. Increase in size plays a dominant role over reduction in stiffness in reducing internalization by macrophages for spherical particles. Internalization of rod-shaped, but not spherical particles, was highly dependent on stiffness. These particles demonstrate the interplay between size, shape and stiffness in interactions of Fc-functionalized particles with macrophages during phagocytosis.
Collapse
Affiliation(s)
- Anusha Garapaty
- School of Chemical & Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA30332
| | - Julie A. Champion
- School of Chemical & Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA30332
| |
Collapse
|
39
|
McCarthy MM, Wright CL. Convergence of Sex Differences and the Neuroimmune System in Autism Spectrum Disorder. Biol Psychiatry 2017; 81:402-410. [PMID: 27871670 PMCID: PMC5285451 DOI: 10.1016/j.biopsych.2016.10.004] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 09/14/2016] [Accepted: 10/04/2016] [Indexed: 01/06/2023]
Abstract
The male bias in autism spectrum disorder incidence is among the most extreme of all neuropsychiatric disorders, yet the origins of the sex difference remain obscure. Developmentally, males are exposed to high levels of testosterone and its byproduct, estradiol. Together these steroids modify the course of brain development by altering neurogenesis, cell death, migration, differentiation, dendritic and axonal growth, synaptogenesis, and synaptic pruning, all of which can be deleteriously impacted during the course of developmental neuropsychiatric disorders. Elucidating the cellular mechanisms by which steroids modulate brain development provides valuable insights into how these processes may go awry. An emerging theme is the role of inflammatory signaling molecules and the innate immune system in directing brain masculinization, the evidence for which we review here. Evidence is also emerging that the neuroimmune system is overactivated in individuals with autism spectrum disorder. These combined observations lead us to propose that the natural process of brain masculinization puts males at risk by moving them closer to a vulnerability threshold that could more easily be breached by inflammation during critical periods of brain development. Two brain regions are highlighted: the preoptic area and the cerebellum. Both are developmentally regulated by the inflammatory prostaglandin E2, but in different ways. Microglia, innate immune cells of the brain, and astrocytes are also critical contributors to masculinization and illustrate the importance of nonneuronal cells to the health of the developing brain.
Collapse
Affiliation(s)
- Margaret M McCarthy
- Department of Pharmacology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland.
| | - Christopher L Wright
- Department of Pharmacology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
40
|
Elizabeth de Sousa Rodrigues M, Bekhbat M, Houser MC, Chang J, Walker DI, Jones DP, Oller do Nascimento CM, Barnum CJ, Tansey MG. Chronic psychological stress and high-fat high-fructose diet disrupt metabolic and inflammatory gene networks in the brain, liver, and gut and promote behavioral deficits in mice. Brain Behav Immun 2017; 59:158-172. [PMID: 27592562 PMCID: PMC5154856 DOI: 10.1016/j.bbi.2016.08.021] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/19/2016] [Accepted: 08/31/2016] [Indexed: 11/25/2022] Open
Abstract
The mechanisms underlying the association between chronic psychological stress, development of metabolic syndrome (MetS), and behavioral impairment in obesity are poorly understood. The aim of the present study was to assess the effects of mild chronic psychological stress on metabolic, inflammatory, and behavioral profiles in a mouse model of diet-induced obesity. We hypothesized that (1) high-fat high-fructose diet (HFHF) and psychological stress would synergize to mediate the impact of inflammation on the central nervous system in the presence of behavioral dysfunction, and that (2) HFHF and stress interactions would impact insulin and lipid metabolism. C57Bl/6 male mice underwent a combination of HFHF and two weeks of chronic psychological stress. MetS-related conditions were assessed using untargeted plasma metabolomics, and structural and immune changes in the gut and liver were evaluated. Inflammation was measured in plasma, liver, gut, and brain. Our results show a complex interplay of diet and stress on gut alterations, energetic homeostasis, lipid metabolism, and plasma insulin levels. Psychological stress and HFHF diet promoted changes in intestinal tight junctions proteins and increases in insulin resistance and plasma cholesterol, and impacted the RNA expression of inflammatory factors in the hippocampus. Stress promoted an adaptive anti-inflammatory profile in the hippocampus that was abolished by diet treatment. HFHF increased hippocampal and hepatic Lcn2 mRNA expression as well as LCN2 plasma levels. Behavioral changes were associated with HFHF and stress. Collectively, these results suggest that diet and stress as pervasive factors exacerbate MetS-related conditions through an inflammatory mechanism that ultimately can impact behavior. This rodent model may prove useful for identification of possible biomarkers and therapeutic targets to treat metabolic syndrome and mood disorders.
Collapse
Affiliation(s)
- Maria Elizabeth de Sousa Rodrigues
- Department of Physiology, School of Medicine at Emory University, United States,Department of Physiology of Nutrition, Federal University of Sao Paulo, SP, Brazil
| | - Mandakh Bekhbat
- Department of Physiology, School of Medicine at Emory University, United States.
| | - Madelyn C. Houser
- Department of Physiology, School of Medicine at Emory University, United States
| | - Jianjun Chang
- Department of Physiology, School of Medicine at Emory University, United States.
| | - Douglas I. Walker
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine at Emory University, United States
| | - Dean P. Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine at Emory University, United States
| | | | | | - Malú G. Tansey
- Department of Physiology, School of Medicine at Emory University, United States,Corresponding author at: Emory University School of Medicine, 605L Whitehead Biomedical Res. Bldg., 615 Michael Street, Atlanta, GA 30322-3110, United States
| |
Collapse
|
41
|
Lawson SK, Gray AC, Woehrle NS. Effects of oxytocin on serotonin 1B agonist-induced autism-like behavior in mice. Behav Brain Res 2016; 314:52-64. [DOI: 10.1016/j.bbr.2016.07.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 07/12/2016] [Accepted: 07/16/2016] [Indexed: 01/19/2023]
|
42
|
Huang H, Nie S, Cao M, Marshall C, Gao J, Xiao N, Hu G, Xiao M. Characterization of AD-like phenotype in aged APPSwe/PS1dE9 mice. AGE (DORDRECHT, NETHERLANDS) 2016; 38:303-322. [PMID: 27439903 PMCID: PMC5061676 DOI: 10.1007/s11357-016-9929-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/12/2016] [Indexed: 05/28/2023]
Abstract
Transgenic APPSwe/PS1dE9 (APP/PS1) mice that overproduce amyloid beta (Aβ) are extensively used in the studies of pathogenesis and experimental therapeutics and new drug screening for Alzheimer's disease (AD). However, most of the current literature uses young or adult APP/PS1 mice. In order to provide a broader view of AD-like phenotype of this animal model, in this study, we systematically analyzed behavioral and pathological profiles of 24-month-old male APP/PS1 mice. Aged APP/PS1 mice had reference memory deficits as well as anxiety, hyperactivity, and social interaction impairment. Consistently, there was obvious deposition of amyloid plaques in the dorsal hippocampus with decreased expression of insulin-degrading enzyme, a proteolytic enzyme responsible for degradation of intracellular Aβ. Furthermore, decreases in hippocampal volume, neuronal number and synaptophysin expression, and astrocyte atrophy were also observed in aged APP/PS1 mice. This finding suggests that aged APP/PS1 mice can well replicate cognitive and noncognitive behavioral abnormalities, hippocampal atrophy, and neuronal and astrocyte degeneration in AD patients, to enable more objective and refined preclinical evaluation of therapeutic drugs and strategies for AD treatment.
Collapse
Affiliation(s)
- Huang Huang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu, 211166, China
- Department of Neurology, Sir Run Run Shaw Hospital, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Sipei Nie
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu, 211166, China
| | - Min Cao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu, 211166, China
| | - Charles Marshall
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu, 211166, China
- Department of Rehabilitation Sciences, University of Kentucky Center of Excellence in Rural Health, Hazard, KY, 41701, USA
| | - Junying Gao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu, 211166, China
| | - Na Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu, 211166, China
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu, 211166, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|