1
|
Birgbauer E. Lysophospholipid receptors in neurodegeneration and neuroprotection. EXPLORATION OF NEUROPROTECTIVE THERAPY 2024; 4:349-365. [PMID: 39247084 PMCID: PMC11379401 DOI: 10.37349/ent.2024.00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/11/2024] [Indexed: 09/10/2024]
Abstract
The central nervous system (CNS) is one of the most complex physiological systems, and treatment of CNS disorders represents an area of major medical need. One critical aspect of the CNS is its lack of regeneration, such that damage is often permanent. The damage often leads to neurodegeneration, and so strategies for neuroprotection could lead to major medical advances. The G protein-coupled receptor (GPCR) family is one of the major receptor classes, and they have been successfully targeted clinically. One class of GPCRs is those activated by bioactive lysophospholipids as ligands, especially sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA). Research has been increasingly demonstrating the important roles that S1P and LPA, and their receptors, play in physiology and disease. In this review, I describe the role of S1P and LPA receptors in neurodegeneration and potential roles in neuroprotection. Much of our understanding of the role of S1P receptors has been through pharmacological tools. One such tool, fingolimod (also known as FTY720), which is a S1P receptor agonist but a functional antagonist in the immune system, is clinically efficacious in multiple sclerosis by producing a lymphopenia to reduce autoimmune attacks; however, there is evidence that fingolimod is also neuroprotective. Furthermore, fingolimod is neuroprotective in many other neuropathologies, including stroke, Parkinson's disease, Huntington's disease, Rett syndrome, Alzheimer's disease, and others that are discussed here. LPA receptors also appear to be involved, being upregulated in a variety of neuropathologies. Antagonists or mutations of LPA receptors, especially LPA1, are neuroprotective in a variety of conditions, including cortical development, traumatic brain injury, spinal cord injury, stroke and others discussed here. Finally, LPA receptors may interact with other receptors, including a functional interaction with plasticity related genes.
Collapse
Affiliation(s)
- Eric Birgbauer
- Department of Biology, Winthrop University, Rock Hill, SC 29733, USA
| |
Collapse
|
2
|
Okabe M, Sato T, Takahashi M, Honjo A, Okawa M, Ishida M, Kukimoto-Niino M, Shirouzu M, Miyamoto Y, Yamauchi J. Autism Spectrum Disorder- and/or Intellectual Disability-Associated Semaphorin-5A Exploits the Mechanism by Which Dock5 Signalosome Molecules Control Cell Shape. Curr Issues Mol Biol 2024; 46:3092-3107. [PMID: 38666924 PMCID: PMC11049140 DOI: 10.3390/cimb46040194] [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: 02/12/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder that includes autism, Asperger's syndrome, and pervasive developmental disorder. Individuals with ASD may exhibit difficulties in social interactions, communication challenges, repetitive behaviors, and restricted interests. While genetic mutations in individuals with ASD can either activate or inactivate the activities of the gene product, impacting neuronal morphogenesis and causing symptoms, the underlying mechanism remains to be fully established. Herein, for the first time, we report that genetically conserved Rac1 guanine-nucleotide exchange factor (GEF) Dock5 signalosome molecules control process elongation in the N1E-115 cell line, a model line capable of achieving neuronal morphological changes. The increased elongation phenotypes observed in ASD and intellectual disability (ID)-associated Semaphorin-5A (Sema5A) Arg676-to-Cys [p.R676C] were also mediated by Dock5 signalosome molecules. Indeed, knockdown of Dock5 using clustered regularly interspaced short palindromic repeat (CRISPR)/CasRx-based guide(g)RNA specifically recovered the mutated Sema5A-induced increase in process elongation in cells. Knockdown of Elmo2, an adaptor molecule of Dock5, also exhibited similar recovery. Comparable results were obtained when transfecting the interaction region of Dock5 with Elmo2. The activation of c-Jun N-terminal kinase (JNK), one of the primary signal transduction molecules underlying process elongation, was ameliorated by either their knockdown or transfection. These results suggest that the Dock5 signalosome comprises abnormal signaling involved in the process elongation induced by ASD- and ID-associated Sema5A. These molecules could be added to the list of potential therapeutic target molecules for abnormal neuronal morphogenesis in ASD at the molecular and cellular levels.
Collapse
Affiliation(s)
- Miyu Okabe
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (M.O.); (Y.M.)
| | - Takanari Sato
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (M.O.); (Y.M.)
| | - Mikito Takahashi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (M.O.); (Y.M.)
| | - Asahi Honjo
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (M.O.); (Y.M.)
| | - Maho Okawa
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (M.O.); (Y.M.)
| | - Miki Ishida
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (M.O.); (Y.M.)
| | - Mutsuko Kukimoto-Niino
- Laboratory for Protein Functional and Structural Biology, Center for Biosystems Dynamics Research, RIKEN, Yokohama 230-0045, Japan; (M.K.-N.); (M.S.)
| | - Mikako Shirouzu
- Laboratory for Protein Functional and Structural Biology, Center for Biosystems Dynamics Research, RIKEN, Yokohama 230-0045, Japan; (M.K.-N.); (M.S.)
| | - Yuki Miyamoto
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (M.O.); (Y.M.)
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (M.O.); (Y.M.)
- Laboratory for Protein Functional and Structural Biology, Center for Biosystems Dynamics Research, RIKEN, Yokohama 230-0045, Japan; (M.K.-N.); (M.S.)
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| |
Collapse
|
3
|
Fukatsu S, Sashi H, Shirai R, Takagi N, Oizumi H, Yamamoto M, Ohbuchi K, Miyamoto Y, Yamauchi J. Rab11a Controls Cell Shape via C9orf72 Protein: Possible Relationships to Frontotemporal Dementia/Amyotrophic Lateral Sclerosis (FTDALS) Type 1. PATHOPHYSIOLOGY 2024; 31:100-116. [PMID: 38390945 PMCID: PMC10885063 DOI: 10.3390/pathophysiology31010008] [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: 12/08/2023] [Revised: 01/24/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024] Open
Abstract
Abnormal nucleotide insertions of C9orf72, which forms a complex with Smith-Magenis syndrome chromosomal region candidate gene 8 (SMCR8) protein and WD repeat-containing protein 41 (WDR41) protein, are associated with an autosomal-dominant neurodegenerative frontotemporal dementia and/or amyotrophic lateral sclerosis type 1 (FTDALS1). The differentially expressed in normal and neoplastic cells (DENN) domain-containing C9orf72 and its complex with SMCR8 and WDR41 function as a guanine-nucleotide exchange factor for Rab GTP/GDP-binding proteins (Rab GEF, also called Rab activator). Among Rab proteins serving as major effectors, there exists Rab11a. However, it remains to be established which Rab protein is related to promoting or sustaining neuronal morphogenesis or homeostasis. In this study, we describe that the knockdown of Rab11a decreases the expression levels of neuronal differentiation marker proteins, as well as the elongation of neurite-like processes, using N1E-115 cells, a well-utilized neuronal differentiation model. Similar results were obtained in primary cortical neurons. In contrast, the knockdown of Rab11b, a Rab11a homolog, did not significantly affect their cell morphological changes. It is of note that treatment with hesperetin, a citrus flavonoid (also known as Vitamin P), recovered the neuronal morphological phenotypes induced by Rab11a knockdown. Also, the knockdown of Rab11a or Rab11b led to a decrease in glial marker expression levels and in morphological changes in FBD-102b cells, which serve as the oligodendroglial differentiation model. Rab11a is specifically involved in the regulation of neuronal morphological differentiation. The knockdown effect mimicking the loss of function of C9orf72 is reversed by treatment with hesperetin. These findings may reveal a clue for identifying one of the potential molecular and cellular phenotypes underlying FTDALS1.
Collapse
Affiliation(s)
- Shoya Fukatsu
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Hinami Sashi
- Laboratory of Applied Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Remina Shirai
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Norio Takagi
- Laboratory of Applied Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Hiroaki Oizumi
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Japan
| | - Masahiro Yamamoto
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Japan
| | - Katsuya Ohbuchi
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Japan
| | - Yuki Miyamoto
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| |
Collapse
|
4
|
Fukatsu S, Okawa M, Okabe M, Cho M, Isogai M, Yokoi T, Shirai R, Oizumi H, Yamamoto M, Ohbuchi K, Miyamoto Y, Yamauchi J. Modulating Golgi Stress Signaling Ameliorates Cell Morphological Phenotypes Induced by CHMP2B with Frontotemporal Dementia-Associated p.Asp148Tyr. Curr Issues Mol Biol 2024; 46:1398-1412. [PMID: 38392208 PMCID: PMC10888485 DOI: 10.3390/cimb46020090] [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: 12/04/2023] [Revised: 01/30/2024] [Accepted: 02/04/2024] [Indexed: 02/24/2024] Open
Abstract
Some charged multivesicular body protein 2B (CHMP2B) mutations are associated with autosomal-dominant neurodegenerative frontotemporal dementia and/or amyotrophic lateral sclerosis type 7 (FTDALS7). The main aim of this study is to clarify the relationship between the expression of mutated CHMP2B protein displaying FTD symptoms and defective neuronal differentiation. First, we illustrate that the expression of CHMP2B with the Asp148Tyr (D148Y) mutation, which preferentially displays FTD phenotypes, blunts neurite process elongation in rat primary cortical neurons. Similar results were observed in the N1E-115 cell line, a model that undergoes neurite elongation. Second, these effects were also accompanied by changes in neuronal differentiation marker protein expression. Third, wild-type CHMP2B protein was indeed localized in the endosomal sorting complexes required to transport (ESCRT)-like structures throughout the cytoplasm. In contrast, CHMP2B with the D148Y mutation exhibited aggregation-like structures and accumulated in the Golgi body. Fourth, among currently known Golgi stress regulators, the expression levels of Hsp47, which has protective effects on the Golgi body, were decreased in cells expressing CHMP2B with the D148Y mutation. Fifth, Arf4, another Golgi stress-signaling molecule, was increased in mutant-expressing cells. Finally, when transfecting Hsp47 or knocking down Arf4 with small interfering (si)RNA, cellular phenotypes in mutant-expressing cells were recovered. These results suggest that CHMP2B with the D148Y mutation, acting through Golgi stress signaling, is negatively involved in the regulation of neuronal cell morphological differentiation, providing evidence that a molecule controlling Golgi stress may be one of the potential FTD therapeutic targets at the molecular and cellular levels.
Collapse
Affiliation(s)
- Shoya Fukatsu
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Maho Okawa
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Miyu Okabe
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Mizuka Cho
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Mikinori Isogai
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Takanori Yokoi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Remina Shirai
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Hiroaki Oizumi
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Japan
| | - Masahiro Yamamoto
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Japan
| | - Katsuya Ohbuchi
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Japan
| | - Yuki Miyamoto
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Setagaya, Tokyo 157-8535, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Setagaya, Tokyo 157-8535, Japan
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-8506, Japan
| |
Collapse
|
5
|
Okabe M, Miyamoto Y, Ikoma Y, Takahashi M, Shirai R, Kukimoto-Niino M, Shirouzu M, Yamauchi J. RhoG-Binding Domain of Elmo1 Ameliorates Excessive Process Elongation Induced by Autism Spectrum Disorder-Associated Sema5A. PATHOPHYSIOLOGY 2023; 30:548-566. [PMID: 38133141 PMCID: PMC10745971 DOI: 10.3390/pathophysiology30040040] [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: 10/10/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder that includes autism, Asperger's syndrome, and pervasive developmental disorder. ASD is characterized by poor interpersonal relationships and strong attachment. The correlations between activated or inactivated gene products, which occur as a result of genetic mutations affecting neurons in ASD patients, and ASD symptoms are now of critical concern. Here, for the first time, we describe the process in which that the respective ASD-associated mutations (Arg676-to-Cys [R676C] and Ser951-to-Cys [S951C]) of semaphorin-5A (Sema5A) localize Sema5A proteins themselves around the plasma membrane in the N1E-115 cell line, a model line that can achieve neuronal morphological differentiation. The expression of each mutated construct resulted in the promotion of excessive elongation of neurite-like processes with increased differentiation protein markers; R676C was more effective than S951C. The differentiated phenotypes were very partially neutralized by an antibody, against Plexin-B3 as the specific Sema5A receptor, suggesting that the effects of Sema5A act in an autocrine manner. R676C greatly increased the activation of c-Jun N-terminal kinase (JNK), one of the signaling molecules underlying process elongation. In contrast, the blocking of JNK signaling, by a chemical JNK inhibitor or an inhibitory construct of the interaction of RhoG with Elmo1 as JNK upstream signaling molecules, recovered the excessive process elongation. These results suggest that ASD-associated mutations of Sema5A, acting through the JNK signaling cascade, lead to excessive differentiated phenotypes, and the inhibition of JNK signaling recovers them, revealing possible therapeutic targets for recovering the potential molecular and cellular phenotypes underlying certain ASD symptoms.
Collapse
Affiliation(s)
- Miyu Okabe
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.M.); (R.S.)
| | - Yuki Miyamoto
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.M.); (R.S.)
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Yuta Ikoma
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.M.); (R.S.)
| | - Mikito Takahashi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.M.); (R.S.)
| | - Remina Shirai
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.M.); (R.S.)
| | - Mutsuko Kukimoto-Niino
- Laboratory for Protein Functional and Structural Biology, Center for Biosystems Dynamics Research, RIKEN, Yokohama 230-0045, Japan (M.S.)
| | - Mikako Shirouzu
- Laboratory for Protein Functional and Structural Biology, Center for Biosystems Dynamics Research, RIKEN, Yokohama 230-0045, Japan (M.S.)
| | - Junji Yamauchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan (Y.M.); (R.S.)
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science,Tokyo 156-8506, Japan
| |
Collapse
|
6
|
Shirai R, Cho M, Isogai M, Fukatsu S, Okabe M, Okawa M, Miyamoto Y, Torii T, Yamauchi J. FTD/ALS Type 7-Associated Thr104Asn Mutation of CHMP2B Blunts Neuronal Process Elongation, and Is Recovered by Knockdown of Arf4, the Golgi Stress Regulator. Neurol Int 2023; 15:980-993. [PMID: 37606396 PMCID: PMC10443297 DOI: 10.3390/neurolint15030063] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/03/2023] [Accepted: 08/10/2023] [Indexed: 08/23/2023] Open
Abstract
Frontotemporal dementia and/or amyotrophic lateral sclerosis type 7 (FTD/ALS7) is an autosomal dominant neurodegenerative disorder characterized by the onset of FTD and/or ALS, mainly in adulthood. Patients with some types of mutations, including the Thr104Asn (T104N) mutation of charged multivesicular body protein 2B (CHMP2B), have predominantly ALS phenotypes, whereas patients with other mutations have predominantly FTD phenotypes. A few mutations result in patients having both phenotypes approximately equally; however, the reason why phenotypes differ depending on the position of the mutation is unknown. CHMP2B comprises one part of the endosomal sorting complexes required for transport (ESCRT), specifically ESCRT-III, in the cytoplasm. We describe here, for the first time, that CHMP2B with the T104N mutation inhibits neuronal process elongation in the N1E-115 cell line, a model line undergoing neuronal differentiation. This inhibitory phenotype was accompanied by changes in marker protein expression. Of note, CHMP2B with the T104N mutation, but not the wild-type form, was preferentially accumulated in the Golgi body. Of the four major Golgi stress signaling pathways currently known, the pathway through Arf4, the small GTPase, was specifically upregulated in cells expressing CHMP2B with the T104N mutation. Conversely, knockdown of Arf4 with the cognate small interfering (si)RNA recovered the neuronal process elongation inhibited by the T104N mutation. These results suggest that the T104N mutation of CHMP2B inhibits morphological differentiation by triggering Golgi stress signaling, revealing a possible therapeutic molecular target for recovering potential molecular and cellular phenotypes underlying FTD/ALS7.
Collapse
Affiliation(s)
- Remina Shirai
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (R.S.); (Y.M.)
| | - Mizuka Cho
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (R.S.); (Y.M.)
| | - Mikinori Isogai
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (R.S.); (Y.M.)
| | - Shoya Fukatsu
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (R.S.); (Y.M.)
| | - Miyu Okabe
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (R.S.); (Y.M.)
| | - Maho Okawa
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (R.S.); (Y.M.)
| | - Yuki Miyamoto
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (R.S.); (Y.M.)
- Department of Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Tomohiro Torii
- Laboratory of Ion Channel Pathophysiology, Doshisha University Graduate School of Brain Science, Kyoto 610-0394, Japan;
| | - Junji Yamauchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (R.S.); (Y.M.)
- Department of Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| |
Collapse
|
7
|
Fukushima N, Shirai R, Sato T, Nakamura S, Ochiai A, Miyamoto Y, Yamauchi J. Knockdown of Rab7B, But Not of Rab7A, Which Antagonistically Regulates Oligodendroglial Cell Morphological Differentiation, Recovers Tunicamycin-Induced Defective Differentiation in FBD-102b Cells. J Mol Neurosci 2023; 73:363-374. [PMID: 37248316 DOI: 10.1007/s12031-023-02117-y] [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: 02/24/2023] [Accepted: 04/06/2023] [Indexed: 05/31/2023]
Abstract
In the central nervous system (CNS), insulative myelin sheaths are generated from the differentiated plasma membranes of oligodendrocytes (oligodendroglial cells) and surround neuronal axons to achieve saltatory conduction. Despite the functional involvement of myelin sheaths in the CNS, the molecular mechanism by which oligodendroglial cells themselves undergo differentiation of plasma membranes remains unclear. It also remains to be explored whether their signaling mechanisms can be applied to treating diseases of the oligodendroglial cells. Here, we describe that Rab7B of Rab7 subfamily small GTPases negatively regulates oligodendroglial cell morphological differentiation using FBD-102b cells, which are model cells undergoing differentiation of oligodendroglial precursors. Knockdown of Rab7B or Rab7A by the respective specific siRNAs in cells positively or negatively regulated morphological differentiation, respectively. Consistently, these changes were supported by changes on differentiation- and myelination-related structural protein and protein kinase markers. We also found that knockdown of Rab7B has the ability to recover inhibition of morphological differentiation following tunicamycin-induced endoplasmic reticulum (ER) stress, which mimics one of the major molecular pathological causes of hereditary hypomyelinating disorders in oligodendroglial cells, such as Pelizaeus-Merzbacher disease (PMD). These results suggest that the respective molecules among very close Rab7 homologues exhibit differential roles in morphological differentiation and that knocking down Rab7B can recover defective differentiating phenotypes under ER stress, thereby adding Rab7B to the list of molecular therapeutic cues taking advantage of signaling mechanisms for oligodendroglial diseases like PMD.
Collapse
Affiliation(s)
- Nana Fukushima
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Remina Shirai
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Takanari Sato
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Sayumi Nakamura
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Arisa Ochiai
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Yuki Miyamoto
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan.
- Department of Pharmacology, National Research Institute for Child Health and Development, 2-10-1, Setagaya, Tokyo, 157-8535, Japan.
| | - Junji Yamauchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan.
- Department of Pharmacology, National Research Institute for Child Health and Development, 2-10-1, Setagaya, Tokyo, 157-8535, Japan.
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo, 156-8506, Japan.
| |
Collapse
|
8
|
Kato Y, Shirai R, Ohbuchi K, Oizumi H, Yamamoto M, Miyata W, Iguchi T, Mimaki Y, Miyamoto Y, Yamauchi J. Hesperetin Ameliorates Inhibition of Neuronal and Oligodendroglial Cell Differentiation Phenotypes Induced by Knockdown of Rab2b, an Autism Spectrum Disorder-Associated Gene Product. Neurol Int 2023; 15:371-391. [PMID: 36976668 PMCID: PMC10057161 DOI: 10.3390/neurolint15010025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/20/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Autism spectrum disorder (ASD) is a central nervous system (CNS) neurodevelopmental disorder that includes autism, pervasive developmental disorder, and Asperger’s syndrome. ASD is characterized by repetitive behaviors and social communication deficits. ASD is thought to be a multifactorial disorder with a range of genetic and environmental factors/candidates. Among such factors is the rab2b gene, although it remains unclear how Rab2b itself is related to the CNS neuronal and glial developmental disorganization observed in ASD patients. Rab2 subfamily members regulate intracellular vesicle transport between the endoplasmic reticulum and the Golgi body. To the best of our knowledge, we are the first to report that Rab2b positively regulates neuronal and glial cell morphological differentiation. Knockdown of Rab2b inhibited morphological changes in N1E-115 cells, which are often used as the neuronal cell differentiation model. These changes were accomplished with decreased expression levels of marker proteins in neuronal cells. Similar results were obtained for FBD-102b cells, which are used as the model of oligodendroglial cell morphological differentiation. In contrast, knockdown of Rab2a, which is another Rab2 family member not known to be associated with ASD, affected only oligodendroglial and not neuronal morphological changes. In contrast, treatment with hesperetin, a citrus flavonoid with various cellular protective effects, in cells recovered the defective morphological changes induced by Rab2b knockdown. These results suggest that knockdown of Rab2b inhibits differentiation in neuronal and glial cells and may be associated with pathological cellular phenotypes in ASD and that hesperetin can recover their phenotypes at the in vitro level at least.
Collapse
Affiliation(s)
- Yukino Kato
- Department of Molecular Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Tokyo, Japan; (Y.K.); (W.M.); (Y.M.)
| | - Remina Shirai
- Department of Molecular Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Tokyo, Japan; (Y.K.); (W.M.); (Y.M.)
| | - Katsuya Ohbuchi
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Ibaraki, Japan; (K.O.); (H.O.); (M.Y.)
| | - Hiroaki Oizumi
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Ibaraki, Japan; (K.O.); (H.O.); (M.Y.)
| | - Masahiro Yamamoto
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Ibaraki, Japan; (K.O.); (H.O.); (M.Y.)
| | - Wakana Miyata
- Department of Molecular Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Tokyo, Japan; (Y.K.); (W.M.); (Y.M.)
| | - Tomoki Iguchi
- Department of Medicinal Pharmacognosy, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Tokyo, Japan; (T.I.); (Y.M.)
| | - Yoshihiro Mimaki
- Department of Medicinal Pharmacognosy, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Tokyo, Japan; (T.I.); (Y.M.)
| | - Yuki Miyamoto
- Department of Molecular Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Tokyo, Japan; (Y.K.); (W.M.); (Y.M.)
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya 157-8535, Tokyo, Japan
| | - Junji Yamauchi
- Department of Molecular Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Tokyo, Japan; (Y.K.); (W.M.); (Y.M.)
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya 157-8535, Tokyo, Japan
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science, Setagaya 156-8506, Tokyo, Japan
- Correspondence: ; Tel.: +81-42-676-7164; Fax: +81-42-676-8841
| |
Collapse
|
9
|
Guan G, Cannon RD, Coates DE, Mei L. Effect of the Rho-Kinase/ROCK Signaling Pathway on Cytoskeleton Components. Genes (Basel) 2023; 14:272. [PMID: 36833199 PMCID: PMC9957420 DOI: 10.3390/genes14020272] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/10/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
The mechanical properties of cells are important in tissue homeostasis and enable cell growth, division, migration and the epithelial-mesenchymal transition. Mechanical properties are determined to a large extent by the cytoskeleton. The cytoskeleton is a complex and dynamic network composed of microfilaments, intermediate filaments and microtubules. These cellular structures confer both cell shape and mechanical properties. The architecture of the networks formed by the cytoskeleton is regulated by several pathways, a key one being the Rho-kinase/ROCK signaling pathway. This review describes the role of ROCK (Rho-associated coiled-coil forming kinase) and how it mediates effects on the key components of the cytoskeleton that are critical for cell behaviour.
Collapse
Affiliation(s)
- Guangzhao Guan
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Department of Oral Diagnostic and Surgical Sciences, Faculty of Dentistry, University of Otago, 310 Great King Street, Dunedin 9016, New Zealand
| | - Richard D. Cannon
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Department of Oral Sciences, Faculty of Dentistry, University of Otago, 310 Great King Street, Dunedin 9016, New Zealand
| | - Dawn E. Coates
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
| | - Li Mei
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Department of Oral Sciences, Faculty of Dentistry, University of Otago, 310 Great King Street, Dunedin 9016, New Zealand
| |
Collapse
|
10
|
Ying X, Yu X, Zhu J, Li X, Zheng Y, Xie Q, Wu Q, Li S, Yue J, Zhou Y, Zhou K, Tu W, Jiang S. Water Treadmill Training Ameliorates Neurite Outgrowth Inhibition Associated with NGR/RhoA/ROCK by Inhibiting Astrocyte Activation following Spinal Cord Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1724362. [PMID: 35387259 PMCID: PMC8977293 DOI: 10.1155/2022/1724362] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 11/19/2021] [Accepted: 03/13/2022] [Indexed: 12/30/2022]
Abstract
Spinal cord injury (SCI) often results in damage to or degeneration of axons. Crosstalk between astrocytes and neurons plays a pivotal role in neurite outgrowth following SCI. Rehabilitative training is a recognized method for the treatment of SCI, but the specific mechanism underlying its effect on axonal outgrowth in the central nervous system (CNS) has not yet been determined. A total of 190 adult male SD rats weighing 200-250 g were randomly divided into eight groups for use as animal models of SCI. Rats were subjected to water treadmill training (TT) for 7 or 14 d. The Basso-Beattie-Bresnahan (BBB) motor function scale, hematoxylin-eosin (HE) staining, Nissl staining, Western blotting, and immunofluorescence were used to measure tissue morphology and the degree of neurological deficit and to determine quantitative expression and accurate localization of the corresponding proteins. We found that TT decreased tissue structure damage and improved functional recovery. TT also promoted the regeneration of neurons and reduced SCI-induced apoptosis SCI around the lesion, as well as significantly increasing the expression of GAP43 and NF200 after SCI. In addition, TT significantly inhibited the injury-induced increase in the expression of proinflammatory factors. Moreover, TT reduced the activation of astrocytes and microglia, accompanied by the reduced expression of C3d and increased expression of S100A10. Finally, TT effectively reduced the level of chondroitin sulfate proteoglycan (CSPG) surrounding the lesion and inhibited the NGR/RhoA/ROCK signaling pathway in neurons after SCI. Overall, we found that TT played a novel role in recovery from SCI by promoting axonal outgrowth associated with NGR/RhoA/ROCK signaling by inhibiting astrocyte activation after SCI.
Collapse
Affiliation(s)
- Xinwang Ying
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
- Department of Intelligent Rehabilitation International (Cross-Strait) Alliance of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Xiaolan Yu
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Jintao Zhu
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Xuqing Li
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Yujun Zheng
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Qingfeng Xie
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Qiaoyun Wu
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Shengcun Li
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
- Department of Intelligent Rehabilitation International (Cross-Strait) Alliance of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Jingjing Yue
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Ye Zhou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), China
| | - Kecheng Zhou
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
- Department of Intelligent Rehabilitation International (Cross-Strait) Alliance of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Wenzhan Tu
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
- Department of Intelligent Rehabilitation International (Cross-Strait) Alliance of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Songhe Jiang
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
- Department of Intelligent Rehabilitation International (Cross-Strait) Alliance of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| |
Collapse
|
11
|
Fujiya T, Asanuma K, Koike T, Okata T, Saito M, Asano N, Imatani A, Masamune A. Nitric oxide could promote development of Barrett's esophagus by S-nitrosylation-induced inhibition of Rho-ROCK signaling in esophageal fibroblasts. Am J Physiol Gastrointest Liver Physiol 2022; 322:G107-G116. [PMID: 34786954 DOI: 10.1152/ajpgi.00124.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Barrett's esophagus arises in the process of wound healing in distal esophageal epithelium damaged by gastroesophageal reflux disease. Differentiation of fibroblast into myofibroblasts, a smooth muscle cell-like phenotype and tissue contraction are crucial processes in wound healing. No study has evaluated mechanism by which luminal esophageal nitric oxide (NO) affect Rho-associated coiled coil-forming protein kinase (Rho-ROCK) signaling pathway, a key factor of tissue contraction, in stromal fibroblasts to develop Barrett's esophagus. Using esophageal fibroblasts, we performed collagen-based cell contraction assays and evaluated influence of Rho-ROCK signaling in the exposure to acidic bile salts and NOC-9, which is an NO donor. We found that enhanced cell contraction induced by acidic bile salts was inhibited by NO, accompanied by decrease in phosphorylated myosin light chain expression and stress fiber formation. NO directly S-nitrosylated GTP-RhoA and consequently blocked Rho-ROCK signaling. Moreover, exposure to NO and Y27632, a Rho-ROCK signaling inhibitor, decreased α-SMA expression and increased bone morphogenetic protein-4 (BMP4) expression and secretion. These findings could account for the increased expression of BMP4 in the columnar epithelial cells and stromal fibroblasts in human Barrett's esophagus. NO could impair wound contraction by blocking the Rho-ROCK signaling pathway and promote the development of Barrett's esophagus.NEW & NOTEWORTHY Barrett's esophagus is the condition where esophageal epithelium damaged by gastroesophageal reflux disease (GERD) is abnormally healed via replacing of metaplastic columnar epithelium, but very few studies have conducted focusing wound healing in the development of Barrett's esophagus. Esophageal luminal nitric oxide inhibits Rho-ROCK signaling pathway in esophageal fibroblasts, which leads to delay tissue contraction, a pivotal step in proper wound healing. Moreover, this inhibition increases tissue BMP4 expression. Impaired wound healing could be related to Barrett's esophagus.
Collapse
Affiliation(s)
- Taku Fujiya
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kiyotaka Asanuma
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomoyuki Koike
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomoki Okata
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masahiro Saito
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naoki Asano
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akira Imatani
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
12
|
Tolomeu HV, Fraga CAM. The Outcomes of Small-Molecule Kinase Inhibitors and the Role of ROCK2 as a Molecular Target for the Treatment of Alzheimer's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 21:188-205. [PMID: 34414875 DOI: 10.2174/1871527320666210820092220] [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: 05/18/2020] [Revised: 02/17/2021] [Accepted: 03/13/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Alzheimer's disease is rapidly becoming a major threat to public health, with an increasing number of individuals affected as the world's population ages. In this sense, studies have been carried out aiming at the identification of new small-molecule kinase inhibitors useful for the treatment of Alzheimer's disease. OBJECTIVE In the present study, we investigated the compounds developed as inhibitors of different protein kinases associated with the pathogenesis of Alzheimer's disease. METHODS The applied methodology was the use of the Clarivate Analytics Integrity and ClinicalTrials. com databases. Moreover, we highlight ROCK2 as a promising target despite being little studied for this purpose. A careful structure-activity relationship analysis of the ROCK2 inhibitors was performed to identify important structural features and fragments for the interaction with the kinase active site, aiming to rationally design novel potent and selective inhibitors. RESULTS We were able to notice some structural characteristics that could serve as the basis to better guide the rational design of new ROCK2 inhibitors as well as some more in-depth characteristics regarding the topology of the active site of both isoforms of these enzymes, thereby identifying differences that could lead to planning more selective compounds. CONCLUSION We hope that this work can be useful to update researchers working in this area, enabling the emergence of new ideas and a greater direction of efforts for designing new ROCK2 inhibitors to identify new therapeutic alternatives for Alzheimer's disease.
Collapse
Affiliation(s)
- Heber Victor Tolomeu
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil | Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941- 902 Rio de Janeiro, RJ, Brazil
| | - Carlos Alberto Manssour Fraga
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil | Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941- 902 Rio de Janeiro, RJ, Brazil
| |
Collapse
|
13
|
Duman JG, Blanco FA, Cronkite CA, Ru Q, Erikson KC, Mulherkar S, Saifullah AB, Firozi K, Tolias KF. Rac-maninoff and Rho-vel: The symphony of Rho-GTPase signaling at excitatory synapses. Small GTPases 2022; 13:14-47. [PMID: 33955328 PMCID: PMC9707551 DOI: 10.1080/21541248.2021.1885264] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 01/15/2023] Open
Abstract
Synaptic connections between neurons are essential for every facet of human cognition and are thus regulated with extreme precision. Rho-family GTPases, molecular switches that cycle between an active GTP-bound state and an inactive GDP-bound state, comprise a critical feature of synaptic regulation. Rho-GTPases are exquisitely controlled by an extensive suite of activators (GEFs) and inhibitors (GAPs and GDIs) and interact with many different signalling pathways to fulfill their roles in orchestrating the development, maintenance, and plasticity of excitatory synapses of the central nervous system. Among the mechanisms that control Rho-GTPase activity and signalling are cell surface receptors, GEF/GAP complexes that tightly regulate single Rho-GTPase dynamics, GEF/GAP and GEF/GEF functional complexes that coordinate multiple Rho-family GTPase activities, effector positive feedback loops, and mutual antagonism of opposing Rho-GTPase pathways. These complex regulatory mechanisms are employed by the cells of the nervous system in almost every step of development, and prominently figure into the processes of synaptic plasticity that underlie learning and memory. Finally, misregulation of Rho-GTPases plays critical roles in responses to neuronal injury, such as traumatic brain injury and neuropathic pain, and in neurodevelopmental and neurodegenerative disorders, including intellectual disability, autism spectrum disorder, schizophrenia, and Alzheimer's Disease. Thus, decoding the mechanisms of Rho-GTPase regulation and function at excitatory synapses has great potential for combatting many of the biggest current challenges in mental health.
Collapse
Affiliation(s)
- Joseph G. Duman
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Francisco A. Blanco
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Integrative Molecular and Biomedical Science Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Christopher A. Cronkite
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Qin Ru
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Kelly C. Erikson
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Shalaka Mulherkar
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Ali Bin Saifullah
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Karen Firozi
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Kimberley F. Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
14
|
Crosas-Molist E, Samain R, Kohlhammer L, Orgaz J, George S, Maiques O, Barcelo J, Sanz-Moreno V. RhoGTPase Signalling in Cancer Progression and Dissemination. Physiol Rev 2021; 102:455-510. [PMID: 34541899 DOI: 10.1152/physrev.00045.2020] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Rho GTPases are a family of small G proteins that regulate a wide array of cellular processes related to their key roles controlling the cytoskeleton. On the other hand, cancer is a multi-step disease caused by the accumulation of genetic mutations and epigenetic alterations, from the initial stages of cancer development when cells in normal tissues undergo transformation, to the acquisition of invasive and metastatic traits, responsible for a large number of cancer related deaths. In this review, we discuss the role of Rho GTPase signalling in cancer in every step of disease progression. Rho GTPases contribute to tumour initiation and progression, by regulating proliferation and apoptosis, but also metabolism, senescence and cell stemness. Rho GTPases play a major role in cell migration, and in the metastatic process. They are also involved in interactions with the tumour microenvironment and regulate inflammation, contributing to cancer progression. After years of intensive research, we highlight the importance of relevant models in the Rho GTPase field, and we reflect on the therapeutic opportunities arising for cancer patients.
Collapse
Affiliation(s)
- Eva Crosas-Molist
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Remi Samain
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Leonie Kohlhammer
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jose Orgaz
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.,Instituto de Investigaciones Biomédicas 'Alberto Sols', CSIC-UAM, 28029, Madrid, Spain
| | - Samantha George
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Oscar Maiques
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jaume Barcelo
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | | |
Collapse
|
15
|
Sampieri L, Funes Chabán M, Di Giusto P, Rozés-Salvador V, Alvarez C. CREB3L2 Modulates Nerve Growth Factor-Induced Cell Differentiation. Front Mol Neurosci 2021; 14:650338. [PMID: 34421533 PMCID: PMC8370844 DOI: 10.3389/fnmol.2021.650338] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 06/24/2021] [Indexed: 11/13/2022] Open
Abstract
Nerve growth factor (NGF) stimulates numerous cellular physiological processes, including growth, differentiation, and survival, and maintains the phenotype of several neuronal types. Most of these NGF-induced processes require adaptation of the secretory pathway since they involve extensive remodeling of membranes and protein redistribution along newly formed neuritic processes. CREB3 transcription factors have emerged as signaling hubs for the regulation of numerous genes involved in the secretory pathway and Golgi homeostasis, integrating stimuli from multiple sources to control secretion, posttranslational modifications and trafficking of proteins. Although recent studies have focused on their role in the central nervous system, little is known about their participation in cell differentiation. Therefore, we aimed to analyze the expression and signaling mechanism of CREB3 transcription factor family members, using the NGF-induced PC12 cell differentiation model. Results show that NGF treatment causes Golgi enlargement and a parallel increased expression of proteins and mRNAs encoding for proteins required for membrane transport (transport factors). Additionally, a significant increase in CREB3L2 protein and mRNA levels is detected in response to NGF. Both MAPK and cAMP signaling pathways are required for this response. Interestingly, CREB3L2 overexpression hampers the NGF-induced neurite outgrowth while its inhibition enhances the morphological changes driven by NGF. In agreement, CREB3L2 overexpressing cells display higher immunofluorescence intensity of Rab5 GTPase (a negative regulator of PC12 differentiation) than control cells. Also, Rab5 immunofluorescence levels decrease in CREB3L2-depleted cells. Taken together, our findings imply that CREB3L2 is an important downstream effector of NGF-activated pathways, leading to neuronal differentiation.
Collapse
Affiliation(s)
- Luciana Sampieri
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Córdoba, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Macarena Funes Chabán
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Córdoba, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Pablo Di Giusto
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Córdoba, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Victoria Rozés-Salvador
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Cecilia Alvarez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Córdoba, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
16
|
Ziegler R, Häusermann F, Kirchner S, Polonchuk L. Cardiac Safety of Kinase Inhibitors - Improving Understanding and Prediction of Liabilities in Drug Discovery Using Human Stem Cell-Derived Models. Front Cardiovasc Med 2021; 8:639824. [PMID: 34222360 PMCID: PMC8242589 DOI: 10.3389/fcvm.2021.639824] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/31/2021] [Indexed: 11/13/2022] Open
Abstract
Many small molecule kinase inhibitors (SMKIs) used to fight cancer have been associated with cardiotoxicity in the clinic. Therefore, preventing their failure in clinical development is a priority for preclinical discovery. Our study focused on the integration and concurrent measurement of ATP, apoptosis dynamics and functional cardiac indexes in human stem cell-derived cardiomyocytes (hSC-CMs) to provide further insights into molecular determinants of compromised cardiac function. Ten out of the fourteen tested SMKIs resulted in a biologically relevant decrease in either beating rate or base impedance (cell number index), illustrating cardiotoxicity as one of the major safety liabilities of SMKIs, in particular of those involved in the PI3K–AKT pathway. Pearson's correlation analysis indicated a good correlation between the different read-outs of functional importance. Therefore, measurement of ATP concentrations and apoptosis in vitro could provide important insight into mechanisms of cardiotoxicity. Detailed investigation of the cellular signals facilitated multi-parameter evaluation allowing integrative assessment of cardiomyocyte behavior. The resulting correlation can be used as a tool to highlight changes in cardiac function and potentially to categorize drugs based on their mechanisms of action.
Collapse
Affiliation(s)
- Ricarda Ziegler
- Pharmaceutical Sciences, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Fabian Häusermann
- Pharmaceutical Sciences, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Stephan Kirchner
- Pharmaceutical Sciences, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Liudmila Polonchuk
- Pharmaceutical Sciences, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
17
|
Liaci C, Camera M, Caslini G, Rando S, Contino S, Romano V, Merlo GR. Neuronal Cytoskeleton in Intellectual Disability: From Systems Biology and Modeling to Therapeutic Opportunities. Int J Mol Sci 2021; 22:ijms22116167. [PMID: 34200511 PMCID: PMC8201358 DOI: 10.3390/ijms22116167] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/25/2021] [Accepted: 06/04/2021] [Indexed: 02/06/2023] Open
Abstract
Intellectual disability (ID) is a pathological condition characterized by limited intellectual functioning and adaptive behaviors. It affects 1–3% of the worldwide population, and no pharmacological therapies are currently available. More than 1000 genes have been found mutated in ID patients pointing out that, despite the common phenotype, the genetic bases are highly heterogeneous and apparently unrelated. Bibliomic analysis reveals that ID genes converge onto a few biological modules, including cytoskeleton dynamics, whose regulation depends on Rho GTPases transduction. Genetic variants exert their effects at different levels in a hierarchical arrangement, starting from the molecular level and moving toward higher levels of organization, i.e., cell compartment and functions, circuits, cognition, and behavior. Thus, cytoskeleton alterations that have an impact on cell processes such as neuronal migration, neuritogenesis, and synaptic plasticity rebound on the overall establishment of an effective network and consequently on the cognitive phenotype. Systems biology (SB) approaches are more focused on the overall interconnected network rather than on individual genes, thus encouraging the design of therapies that aim to correct common dysregulated biological processes. This review summarizes current knowledge about cytoskeleton control in neurons and its relevance for the ID pathogenesis, exploiting in silico modeling and translating the implications of those findings into biomedical research.
Collapse
Affiliation(s)
- Carla Liaci
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Mattia Camera
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Giovanni Caslini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Simona Rando
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Salvatore Contino
- Department of Engineering, University of Palermo, Viale delle Scienze Ed. 8, 90128 Palermo, Italy;
| | - Valentino Romano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze Ed. 16, 90128 Palermo, Italy;
| | - Giorgio R. Merlo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
- Correspondence: ; Tel.: +39-0116706449; Fax: +39-0116706432
| |
Collapse
|
18
|
Birgbauer E. Lysophosphatidic Acid Signalling in Nervous System Development and Function. Neuromolecular Med 2021; 23:68-85. [PMID: 33151452 PMCID: PMC11420905 DOI: 10.1007/s12017-020-08630-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/30/2020] [Indexed: 02/06/2023]
Abstract
One class of molecules that are now coming to be recognized as essential for our understanding of the nervous system are the lysophospholipids. One of the major signaling lysophospholipids is lysophosphatidic acid, also known as LPA. LPA activates a variety of G protein-coupled receptors (GPCRs) leading to a multitude of physiological responses. In this review, I describe our current understanding of the role of LPA and LPA receptor signaling in the development and function of the nervous system, especially the central nervous system (CNS). In addition, I highlight how aberrant LPA receptor signaling may underlie neuropathological conditions, with important clinical application.
Collapse
Affiliation(s)
- Eric Birgbauer
- Department of Biology, Winthrop University, Rock Hill, SC, USA.
| |
Collapse
|
19
|
Horváth ÁI, Gyimesi M, Várkuti BH, Képiró M, Szegvári G, Lőrincz I, Hegyi G, Kovács M, Málnási-Csizmadia A. Effect of allosteric inhibition of non-muscle myosin 2 on its intracellular diffusion. Sci Rep 2020; 10:13341. [PMID: 32769996 PMCID: PMC7415145 DOI: 10.1038/s41598-020-69853-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 07/03/2020] [Indexed: 12/12/2022] Open
Abstract
Subcellular dynamics of non-muscle myosin 2 (NM2) is crucial for a broad-array of cellular functions. To unveil mechanisms of NM2 pharmacological control, we determined how the dynamics of NM2 diffusion is affected by NM2′s allosteric inhibitors, i.e. blebbistatin derivatives, as compared to Y-27632 inhibiting ROCK, NM2′s upstream regulator. We found that NM2 diffusion is markedly faster in central fibers than in peripheral stress fibers. Y-27632 accelerated NM2 diffusion in both peripheral and central fibers, whereas in peripheral fibers blebbistatin derivatives slightly accelerated NM2 diffusion at low, but markedly slowed it at high inhibitor concentrations. In contrast, rapid NM2 diffusion in central fibers was unaffected by direct NM2 inhibition. Using our optopharmacological tool, Molecular Tattoo, sub-effective concentrations of a photo-crosslinkable blebbistatin derivative were increased to effective levels in a small, irradiated area of peripheral fibers. These findings suggest that direct allosteric inhibition affects the diffusion profile of NM2 in a markedly different manner compared to the disruption of the upstream control of NM2. The pharmacological action of myosin inhibitors is channeled through autonomous molecular processes and might be affected by the load acting on the NM2 proteins.
Collapse
Affiliation(s)
- Ádám I Horváth
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c, 1117, Budapest, Hungary
| | - Máté Gyimesi
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c, 1117, Budapest, Hungary
| | - Boglárka H Várkuti
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c, 1117, Budapest, Hungary
| | - Miklós Képiró
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c, 1117, Budapest, Hungary
| | - Gábor Szegvári
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c, 1117, Budapest, Hungary
| | - István Lőrincz
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c, 1117, Budapest, Hungary
| | - György Hegyi
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c, 1117, Budapest, Hungary
| | - Mihály Kovács
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c, 1117, Budapest, Hungary.
| | - András Málnási-Csizmadia
- MTA-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c, 1117, Budapest, Hungary.
| |
Collapse
|
20
|
Ito H, Yamashita Y, Tanaka T, Takaki M, Le MN, Yoshida LM, Morimoto K. Cigarette smoke induces endoplasmic reticulum stress and suppresses efferocytosis through the activation of RhoA. Sci Rep 2020; 10:12620. [PMID: 32724133 PMCID: PMC7387437 DOI: 10.1038/s41598-020-69610-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 07/15/2020] [Indexed: 12/22/2022] Open
Abstract
Impaired efferocytosis is a key mechanism of inflammatory lung diseases, including chronic obstructive pulmonary disease and cystic fibrosis. Cigarette smoking activates RhoA and impairs efferocytosis in alveolar macrophages, but the mechanism has not been fully elucidated. We investigated the role of endoplasmic reticulum (ER) stress induced by cigarette smoking in the disruption of efferocytosis. Both tunicamycin (10 μg/ml) and thapsigargin (0.1 and 1 μM), which are ER stress inducers, suppressed efferocytosis in J774 cells, and a Rho-associated coiled-coil-forming kinase (ROCK) inhibitor (Y27632) reversed this effect. We validated the effect of tunicamycin on efferocytosis in experiments using RAW264.7 cells. Then, we investigated the role of the unfolded protein response (UPR) in efferocytosis impaired by ER stress. A PERK inhibitor (GSK2606414) restored the efferocytosis that had been impaired by TM, and an eIF2α dephosphorylation inhibitor (salubrinal) suppressed efferocytosis. Cigarette smoke extract (CSE) induced ER stress in J774 macrophages and RhoA activation in J774 cells, and the CSE-induced ROCK activity was successfully reversed by GSK2606414 and tauroursodeoxycholic acid. Finally, we confirmed that ER stress suppresses efferocytosis in murine alveolar macrophages and that GSK2606414 could rescue this process. These data suggest that cigarette smoke-induced ER stress and the UPR play crucial roles in RhoA activation and suppression of efferocytosis in the lung.
Collapse
Affiliation(s)
- Hiroyuki Ito
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki City, Nagasaki, 852-8523, Japan.,Department of Clinical Tropical Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yoshiro Yamashita
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki City, Nagasaki, 852-8523, Japan
| | - Takeshi Tanaka
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki City, Nagasaki, 852-8523, Japan
| | - Masahiro Takaki
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki City, Nagasaki, 852-8523, Japan
| | - Minh Nhat Le
- Department of Pediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Lay-Myint Yoshida
- Department of Pediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Konosuke Morimoto
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki City, Nagasaki, 852-8523, Japan. .,Department of Clinical Tropical Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| |
Collapse
|
21
|
Maternal Dietary Exposure to Low-Dose Bisphenol A Affects Metabolic and Signaling Pathways in the Brain of Rat Fetuses. Nutrients 2020; 12:nu12051448. [PMID: 32429515 PMCID: PMC7285067 DOI: 10.3390/nu12051448] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/08/2020] [Accepted: 05/14/2020] [Indexed: 12/23/2022] Open
Abstract
Bisphenol A (BPA) is a synthetic compound widely used for the production of polycarbonate plasticware and epoxy resins. BPA exposure is widespread and more than 90% of individuals have detectable amounts of the molecule in their body fluids, which originates primarily from diet. Here, we investigated whether prenatal exposure to BPA affects the mevalonate (MVA) pathway in rat brain fetuses, and whether potential effects are sex-dependent. The MVA pathway is important for brain development and function. Our results demonstrate that the fetal brain, exposed in utero to a very low dose of BPA (2.5 µg/kg/day), displayed altered MVA pathway activation, increased protein prenylation, and a decreased level of pro-BDNF. Interestingly, the BPA-induced effects on estrogen receptor α were sex-dependent. In conclusion, this work demonstrates intergenerational effects of BPA on the brain at very low doses. Our results reveal new targets for BPA-induced interference and underline the impacts of BPA on health.
Collapse
|
22
|
Sin WC, Tam N, Moniz D, Lee C, Church J. Na/H exchanger NHE1 acts upstream of rho GTPases to promote neurite outgrowth. J Cell Commun Signal 2020; 14:325-333. [PMID: 32144636 DOI: 10.1007/s12079-020-00556-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/28/2020] [Indexed: 02/05/2023] Open
Abstract
Na+/H+ exchanger NHE1, a major determinant of intracellular pH (pHi) in mammalian central neurons, promotes neurite outgrowth under both basal and netrin-1-stimulated conditions. The small GTP binding proteins and their effectors have a dominant role in netrin-1-stimulated neurite outgrowth. Since NHE1 has been shown previously to work downstream of the Rho GTPases-mediated polarized membrane protrusion in non-neuronal cells, we examined whether NHE1 has a similar relationship with Cdc42, Rac1 and RhoA in neuronal morphogenesis. Interestingly, our results suggest the possibility that NHE1 acting upstream of Rho GTPases to promote neurite outgrowth induced by netrin-1. First, we found that netrin-1-induced increases in the activities of Rho GTPases using FRET (Forster Resonance Energy Transfer) analyses in individual growth cones; furthermore, their increased activities were abolished by cariporide, a specific NHE1 inhibitor. Second, NHE1 inhibition had no effect on neurite retraction induced by L-α-Lysophosphatidic acid (LPA), a potent RhoA activator. The regulation of Rho GTPases by NHE1 was further evidenced by reduced Rac1, Cdc42 and RhoA activities in NHE1-null neurons. Taken together, our findings suggest that NHE1-dependent neuronal morphogenesis involves the activation of Rho-family of small GTPases.
Collapse
Affiliation(s)
- Wun Chey Sin
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada.
| | - Nicola Tam
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - David Moniz
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Connie Lee
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - John Church
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
23
|
Wypych D, Pomorski P. Calcium Signaling in Glioma Cells: The Role of Nucleotide Receptors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1202:67-86. [PMID: 32034709 DOI: 10.1007/978-3-030-30651-9_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Calcium signaling is probably one of the evolutionary oldest and the most common way by which the signal can be transmitted from the cell environment to the cytoplasmic calcium binding effectors. Calcium signal is fast and due to diversity of calcium binding proteins it may have a very broad effect on cell behavior. Being a crucial player in neuronal transmission it is also very important for glia physiology. It is responsible for the cross-talk between neurons and astrocytes, for microglia activation and motility. Changes in calcium signaling are also crucial for the behavior of transformed glioma cells. The present chapter summarizes molecular mechanisms of calcium signal formation present in glial cells with a strong emphasis on extracellular nucleotide-evoked signaling pathways. Some aspects of glioma C6 signaling such as the cross-talk between P2Y1 and P2Y12 nucleotide receptors in calcium signal generation will be discussed in-depth, to show complexity of machinery engaged in formation of this signal. Moreover, possible mechanisms of modulation of the calcium signal in diverse environments there will be presented herein. Finally, the possible role of calcium signal in glioma motility is also discussed. This is a very important issue, since glioma cells, contrary to the vast majority of neoplastic cells, cannot spread in the body with the bloodstream and, at least in early stages of tumor development, may expand only by means of sheer motility.
Collapse
Affiliation(s)
- Dorota Wypych
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Paweł Pomorski
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
24
|
Sharma P, Roy K. ROCK-2-selective targeting and its therapeutic outcomes. Drug Discov Today 2020; 25:446-455. [DOI: 10.1016/j.drudis.2019.11.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/08/2019] [Accepted: 11/30/2019] [Indexed: 01/21/2023]
|
25
|
Mir SS, Bhat HF, Bhat ZF. Dynamic actin remodeling in response to lysophosphatidic acid. J Biomol Struct Dyn 2020; 38:5253-5265. [PMID: 31920158 DOI: 10.1080/07391102.2019.1696230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Lysophosphatidic acid (LPA) is a multifunctional regulator of actin cytoskeleton that exerts a dramatic impact on the actin cytoskeleton to build a platform for diverse cellular processes including growth cone guidance, neurite retraction and cell motility. It has been implicated in the formation and dissociation of complexes between actin and actin binding proteins, supporting its role in actin remodeling. Several studies point towards its ability to facilitate formation of special cellular structures including focal adhesions and actin stress fibres by phosphoregulation of several actin associated proteins and their multiple regulatory kinases and phosphatases. In addition, multiple levels of crosstalk among the signaling cascades activated by LPA, affect actin cytoskeleton-mediated cell migration and chemotaxis which in turn play a crucial role in cancer metastasis. In the current review, we have attempted to highlight the role of LPA as an actin modulator which functions by controlling activities of specific cellular proteins that underlie mechanisms employed in cytoskeletal and pathophysiological events within the cell. Further studies on the actin affecting/remodeling activity of LPA in different cell types will no doubt throw up many surprises essential to gain a full understanding of its contribution in physiological processes as well as in diseases.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Saima S Mir
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu And Kashmir, India.,Division of Animal Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, Jammu And Kashmir, India
| | - Hina F Bhat
- Division of Animal Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, Jammu And Kashmir, India
| | - Zuhaib F Bhat
- Department of Wine, Food & Molecular Biosciences, Lincoln University, Lincoln, New Zealand.,Division of Livestock Products and Technology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu (SKUAST-J), R.S. Pora, Jammu And Kashmir, India
| |
Collapse
|
26
|
Javier-Torrent M, Marco S, Rocandio D, Pons-Vizcarra M, Janes PW, Lackmann M, Egea J, Saura CA. Presenilin/γ-secretase-dependent EphA3 processing mediates axon elongation through non-muscle myosin IIA. eLife 2019; 8:43646. [PMID: 31577226 PMCID: PMC6774734 DOI: 10.7554/elife.43646] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 09/18/2019] [Indexed: 01/08/2023] Open
Abstract
EphA/ephrin signaling regulates axon growth and guidance of neurons, but whether this process occurs also independently of ephrins is unclear. We show that presenilin-1 (PS1)/γ-secretase is required for axon growth in the developing mouse brain. PS1/γ-secretase mediates axon growth by inhibiting RhoA signaling and cleaving EphA3 independently of ligand to generate an intracellular domain (ICD) fragment that reverses axon defects in PS1/γ-secretase- and EphA3-deficient hippocampal neurons. Proteomic analysis revealed that EphA3 ICD binds to non-muscle myosin IIA (NMIIA) and increases its phosphorylation (Ser1943), which promotes NMIIA filament disassembly and cytoskeleton rearrangement. PS1/γ-secretase-deficient neurons show decreased phosphorylated NMIIA and NMIIA/actin colocalization. Moreover, pharmacological NMII inhibition reverses axon retraction in PS-deficient neurons suggesting that NMIIA mediates PS/EphA3-dependent axon elongation. In conclusion, PS/γ-secretase-dependent EphA3 cleavage mediates axon growth by regulating filament assembly through RhoA signaling and NMIIA, suggesting opposite roles of EphA3 on inhibiting (ligand-dependent) and promoting (receptor processing) axon growth in developing neurons.
Collapse
Affiliation(s)
- Míriam Javier-Torrent
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sergi Marco
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Daniel Rocandio
- Institut de Recerca Biomédica de Lleida, Universitat de Lleida, Lleida, Spain
| | - Maria Pons-Vizcarra
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Peter W Janes
- Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Martin Lackmann
- Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Joaquim Egea
- Institut de Recerca Biomédica de Lleida, Universitat de Lleida, Lleida, Spain
| | - Carlos A Saura
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
27
|
Hu Y, Li X, Huang G, Wang J, Lu W. Fasudil may induce the differentiation of bone marrow mesenchymal stem cells into neuron‑like cells via the Wnt/β‑catenin pathway. Mol Med Rep 2019; 19:3095-3104. [PMID: 30816472 PMCID: PMC6423592 DOI: 10.3892/mmr.2019.9978] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 02/18/2019] [Indexed: 01/27/2023] Open
Abstract
Bone mesenchymal stem cells (MSCs) are an excellent donor graft source due to their potential for self-renewal and multidirectional differentiation. However, it is difficult to obtain high quality MSCs and to induce them to differentiate into neuron-like cells. Fasudil, a Rho kinase inhibitor, exhibits therapeutic potential in spinal cord injuries and stroke. The present study investigated the effect of fasudil on the differentiation of MSCs into neuron-like cells. MSCs were obtained from rat femur marrow, expanded in culture medium, and used at the third passage for subsequent experiments. MSCs were pre-induced with 10 ng/ml basic fibroblast growth factor (bFGF) for 24 h, which was followed by induction with fasudil. A control untreated group and a group treated with fasudil + XAV939, a Wnt/β-catenin pathway inhibitor, were also used in the present study. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR), western blot analysis and immunofluorescence staining were performed in order to detect neuron-specific markers, including neuron-specific enolase (NSE), nestin and neurofilament-M (NF-M). Following induction with fasudil, neuron-like cell morphology was observed. In the fasudil + XAV939 and control groups, no obvious changes in cell shape were observed. The results of RT-qPCR, western blot analysis and immunofluorescence staining indicated that expression of the neuron-specific markers NSE, nestin and NF-M was detected in the fasudil group. The differentiation of MSCs into neuron-like cells induced by fasudil was eliminated when the Wnt/β-catenin pathway was inhibited. The present study demonstrated that fasudil may induce MSCs to differentiate into neuron-like cells, however further studies are required to determine the specific mechanisms involved in the effect of fasudil on the Wnt/β-catenin pathway. In addition, further research is required to examine the functional characteristics of the induced neuron-like cells, in order to establish their suitability for clinical treatments in the future.
Collapse
Affiliation(s)
- Yahui Hu
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Xin Li
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Guowei Huang
- Department of Nutrition and Food Hygiene, School of Public Health, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Jizuo Wang
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Wei Lu
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| |
Collapse
|
28
|
Wang J, Sui RX, Miao Q, Wang Q, Song LJ, Yu JZ, Li YH, Xiao BG, Ma CG. Hydroxyfasudil alleviates demyelination through the inhibition of MOG antibody and microglia activation in cuprizone mouse model. Clin Immunol 2019; 201:35-47. [PMID: 30660624 DOI: 10.1016/j.clim.2019.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 01/14/2019] [Accepted: 01/14/2019] [Indexed: 02/06/2023]
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system characterized by oligodendrocyte loss and progressive neurodegeneration. The cuprizone (CPZ)-induced demyelination is widely used to investigate the demyelination/remyelination. Here, we explored the therapeutic effects of Hydroxyfasudil (HF), an active metabolite of Fasudil, in CPZ model. HF improved behavioral abnormality and reduced myelin damage in the corpus callosum. Splenic atrophy and myelin oligodendrocyte glycoprotein (MOG) antibody were observed in CPZ model, which were partially restored and obviously inhibited by HF, therefore reducing pathogenic binding of MOG antibody to oligodendrocytes. HF inhibited the percentages of CD4+IL-17+ T cells from splenocytes and infiltration of CD4+ T cells and CD68+ macrophages in the brain. HF also declined microglia-mediated neuroinflammation, and promoted the production of astrocyte-derived brain derived neurotrophic factor (BDNF) and regeneration of NG2+ oligodendrocyte precursor cells. These results provide potent evidence for the therapeutic effects of HF in CPZ-induced demyelination.
Collapse
Affiliation(s)
- Jing Wang
- Shanxi Medical University, Taiyuan030001, China
| | - Ruo-Xuan Sui
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan030024, China
| | - Qiang Miao
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan030024, China
| | - Qing Wang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan030024, China
| | - Li-Juan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan030024, China
| | - Jie-Zhong Yu
- Institute of Brain Science, Shanxi Datong University, Datong037009, China
| | - Yan-Hua Li
- Institute of Brain Science, Shanxi Datong University, Datong037009, China
| | - Bao-Guo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200025, China.
| | - Cun-Gen Ma
- Shanxi Medical University, Taiyuan030001, China; The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan030024, China; Institute of Brain Science, Shanxi Datong University, Datong037009, China.
| |
Collapse
|
29
|
Greathouse KM, Boros BD, Deslauriers JF, Henderson BW, Curtis KA, Gentry EG, Herskowitz JH. Distinct and complementary functions of rho kinase isoforms ROCK1 and ROCK2 in prefrontal cortex structural plasticity. Brain Struct Funct 2018; 223:4227-4241. [PMID: 30196430 PMCID: PMC6252131 DOI: 10.1007/s00429-018-1748-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 09/04/2018] [Indexed: 12/13/2022]
Abstract
Rho-associated protein kinases (ROCK) 1 and 2 are attractive drug targets for a range of neurologic disorders; however, a critical barrier to ROCK-based therapeutics is ambiguity over whether there are isoform-specific roles for ROCKs in neuronal structural plasticity. Here, we used a genetics approach to address this long-standing question by analyzing both male and female adult ROCK1+/- and ROCK2+/- mice compared to littermate controls. Individual pyramidal neurons in the medial prefrontal cortex (mPFC) were targeted for iontophoretic microinjection of fluorescent dye, followed by high-resolution confocal microscopy and neuronal 3D reconstructions for morphometry analysis. Increased apical and basal dendritic length and intersections were observed in ROCK1+/- but not ROCK2+/- mice. Although dendritic spine densities were comparable among genotypes, apical spine length was decreased in ROCK1+/- but increased in ROCK2+/- mice. Spine head and neck diameter were reduced similarly in ROCK1+/- and ROCK2+/- mice; however, certain spine morphologic subclasses were more affected than others in a genotype-dependent manner. Biochemical analyses of ROCK substrates in synaptic fractions revealed that phosphorylation of LIM kinase and cofilin were reduced in ROCK1+/- and ROCK2+/- mice, while phosphorylation of myosin light chain was decreased exclusively in ROCK1+/- mice. Collectively, these observations implicate ROCK1 as a novel regulatory factor of neuronal dendritic structure and detail distinct and complementary roles of ROCKs in mPFC dendritic spine structure.
Collapse
Affiliation(s)
- Kelsey M Greathouse
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, 35294, USA
| | - Benjamin D Boros
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, 35294, USA
| | - Josue F Deslauriers
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, 35294, USA
| | - Benjamin W Henderson
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, 35294, USA
| | - Kendall A Curtis
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, 35294, USA
| | - Erik G Gentry
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, 35294, USA
| | - Jeremy H Herskowitz
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, 35294, USA.
| |
Collapse
|
30
|
Functions of Rhotekin, an Effector of Rho GTPase, and Its Binding Partners in Mammals. Int J Mol Sci 2018; 19:ijms19072121. [PMID: 30037057 PMCID: PMC6073136 DOI: 10.3390/ijms19072121] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 12/26/2022] Open
Abstract
Rhotekin is an effector protein for small GTPase Rho. This protein consists of a Rho binding domain (RBD), a pleckstrin homology (PH) domain, two proline-rich regions and a C-terminal PDZ (PSD-95, Discs-large, and ZO-1)-binding motif. We, and other groups, have identified various binding partners for Rhotekin and carried out biochemical and cell biological characterization. However, the physiological functions of Rhotekin, per se, are as of yet largely unknown. In this review, we summarize known features of Rhotekin and its binding partners in neuronal tissues and cancer cells.
Collapse
|
31
|
Sabbir MG, Calcutt NA, Fernyhough P. Muscarinic Acetylcholine Type 1 Receptor Activity Constrains Neurite Outgrowth by Inhibiting Microtubule Polymerization and Mitochondrial Trafficking in Adult Sensory Neurons. Front Neurosci 2018; 12:402. [PMID: 29997469 PMCID: PMC6029366 DOI: 10.3389/fnins.2018.00402] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/24/2018] [Indexed: 11/13/2022] Open
Abstract
The muscarinic acetylcholine type 1 receptor (M1R) is a metabotropic G protein-coupled receptor. Knockout of M1R or exposure to selective or specific receptor antagonists elevates neurite outgrowth in adult sensory neurons and is therapeutic in diverse models of peripheral neuropathy. We tested the hypothesis that endogenous M1R activation constrained neurite outgrowth via a negative impact on the cytoskeleton and subsequent mitochondrial trafficking. We overexpressed M1R in primary cultures of adult rat sensory neurons and cell lines and studied the physiological and molecular consequences related to regulation of cytoskeletal/mitochondrial dynamics and neurite outgrowth. In adult primary neurons, overexpression of M1R caused disruption of the tubulin, but not actin, cytoskeleton and significantly reduced neurite outgrowth. Over-expression of a M1R-DREADD mutant comparatively increased neurite outgrowth suggesting that acetylcholine released from cultured neurons interacts with M1R to suppress neurite outgrowth. M1R-dependent constraint on neurite outgrowth was removed by selective (pirenzepine) or specific (muscarinic toxin 7) M1R antagonists. M1R-dependent disruption of the cytoskeleton also diminished mitochondrial abundance and trafficking in distal neurites, a disorder that was also rescued by pirenzepine or muscarinic toxin 7. M1R activation modulated cytoskeletal dynamics through activation of the G protein (Gα13) that inhibited tubulin polymerization and thus reduced neurite outgrowth. Our study provides a novel mechanism of M1R control of Gα13 protein-dependent modulation of the tubulin cytoskeleton, mitochondrial trafficking and neurite outgrowth in axons of adult sensory neurons. This novel pathway could be harnessed to treat dying-back neuropathies since anti-muscarinic drugs are currently utilized for other clinical conditions.
Collapse
Affiliation(s)
- Mohammad G Sabbir
- Division of Neurodegenerative Disorders, St. Boniface Hospital Research Centre, Winnipeg, MB, Canada
| | - Nigel A Calcutt
- Department of Pathology, University of California, San Diego, San Diego, CA, United States
| | - Paul Fernyhough
- Division of Neurodegenerative Disorders, St. Boniface Hospital Research Centre, Winnipeg, MB, Canada.,Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
32
|
Narumiya S, Thumkeo D. Rho signaling research: history, current status and future directions. FEBS Lett 2018; 592:1763-1776. [PMID: 29749605 PMCID: PMC6032899 DOI: 10.1002/1873-3468.13087] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 12/24/2022]
Abstract
One of the main research areas in biology from the mid‐1980s through the 1990s was the elucidation of signaling pathways governing cell responses. These studies brought, among other molecules, the small GTPase Rho to the epicenter. Rho signaling research has since expanded to all areas of biology and medicine. Here, we describe how Rho emerged as a key molecule governing cell morphogenesis and movement, how it was linked to actin reorganization, and how the study of Rho signaling has expanded from cultured cells to whole biological systems. We then give an overview of the current research status of Rho signaling in development, brain, cardiovascular system, immunity and cancer, and discuss the future directions of Rho signaling research, with emphasis on one Rho effector, ROCK*.
*The Rho GTPase family. Rho family GTPases have now expanded to contain 20 members. Amino acid sequences of 20 Rho GTPases found in human were aligned and the phylogenetic tree was generated by ClustalW2 software (EMBL‐EBI) based on NJ algorithm. The subfamilies of the Rho GTPases are highlighted by the circle and labeled on the right side. Rho cited in this review refers to the original members of Rho subfamily, RhoA, RhoB and RhoC, that are C3 substrates, and, unless specified, not to other members of the Rho subfamily such as Rac, Cdc42, and Rnd. ![]()
Collapse
Affiliation(s)
- Shuh Narumiya
- Department of Drug Discovery Medicine, Medical Innovation Center, Kyoto University Graduate School of Medicine, Japan
| | - Dean Thumkeo
- Department of Drug Discovery Medicine, Medical Innovation Center, Kyoto University Graduate School of Medicine, Japan
| |
Collapse
|
33
|
Zhao Z, Hu J, Gao X, Liang H, Yu H, Liu S, Liu Z. Hyperglycemia via activation of thromboxane A2 receptor impairs the integrity and function of blood-brain barrier in microvascular endothelial cells. Oncotarget 2018; 8:30030-30038. [PMID: 28415790 PMCID: PMC5444723 DOI: 10.18632/oncotarget.16273] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 03/06/2017] [Indexed: 01/02/2023] Open
Abstract
Diabetes is one of high risk factors for cardio- and cerebra-vascular diseases, including stroke, atherosclerosis and hypertension. This study was conducted to elucidate whether and how thromboxane receptor (TPr) activation contributes to blood-brain barrier (BBB) dysfunction in diabetes. Human brain microvascular endothelial cells (HBMECs) were cultured. The levels of phosphorylated endothelial nitric oxide synthase (eNOS) at Ser1177 (p-eNOS) and Akt at Ser473 (p-Akt) were assayed by western blot. Exposure of HBMECs to either high glucose (HG) or thromboxane A2 (TxA2) mimetic U46619, significantly reduced p-eNOS and p-Akt. These effects were abolished by pharmacological or genetic inhibitors of TPr. HG/U46619-induced suppressions of eNOS and Akt phosphorylation were accompanied by upregulation of PTEN and Ser380/Thr382/383 PTEN phosphorylation. PTEN-specific siRNA restored Akt-eNOS signaling in the face of TPr activation or HG. The small GTPase, Rho, was also activated by HG stimulation, and pretreatment of HBMECs with Y27632, a Rho-associated kinase (ROCK) inhibitor, rescued HG-impaired Akt-eNOS signaling. In STZ-injected rats, we found that hyperglycemia dramatically increased the levels of PTEN and PTEN-Ser380/Thr382/383 phosphorylation, reduced both levels of p-eNOS and p-Akt, and disrupted BBB function assayed by Evans blue staining, which were abolished by SQ29548 treatment. We conclude that hyperglycemia activates thromboxane A2 receptor to impair the integrity and function of blood-brain barrier via the ROCK-PTEN-Akt-eNOS pathway.
Collapse
Affiliation(s)
- Zhihong Zhao
- Department of Neurology, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| | - Jue Hu
- Department of Neurology, Changsha Central Hospital, Changsha, Hunan, China
| | - Xiaoping Gao
- Department of Neurology, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| | - Hui Liang
- Department of Neurology, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| | - Haiya Yu
- Department of Neurology, The People's Hospital of Xishui, Huangang, Hubei, China
| | - Suosi Liu
- Department of Neurology, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China.,Department of Clinical Nutrition and Gastroenterology, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| | - Zhan Liu
- Department of Clinical Nutrition and Gastroenterology, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
34
|
Zhang X, Wu N. Fasudil inhibits proliferation and migration of Hep-2 laryngeal carcinoma cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:373-381. [PMID: 29503530 PMCID: PMC5825979 DOI: 10.2147/dddt.s147547] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Rho-kinase signal pathway is a new target for cancer therapy. Fasudil, a selective Rho-kinase inhibitor, is found to exert antitumor effects on several types of cancer, but whether fasudil has antitumor effects on laryngeal carcinoma is still unknown. The aim of this study was to determine the effects of fasudil on laryngeal carcinoma and explore the underlying molecular mechanisms in this process. Methods After treatment with fasudil, changes in biological behaviors, including the growth, proliferation, clone formation, apoptosis, and migration of human laryngeal carcinoma cells (Hep-2 cells) were observed. The influences on apoptotic protease activity factor-1 (APAF-1)-mediated apoptosis pathway and the activities of matrix metalloproteinases (MMP-2 and MMP-9) were measured by Western blotting and gelatin zymography assay. Results Half-maximal inhibitory concentration of fasudil to Hep-2 cells was ~3.40×103 µM (95% CI: 2.53-4.66×103 µM). Moreover, fasudil treatment significantly decreased the ability of growth, proliferation, clone formation, and migration of Hep-2 cells, while remarkably increased the apoptosis rate. Furthermore, the expressions of APAF-1, caspase-9, and caspase-3 significantly increased in fasudil treatment group. Meanwhile, fasudil led to a remarkable decrease in the expressions and activities of MMP-2 and MMP-9. Conclusion Our findings first demonstrate that fasudil not only inhibits the proliferation of laryngeal carcinoma cells through activating APAF-1-mediated apoptosis pathway, but also prevents migration by inhibiting the activities of MMP-2 and MMP-9. Therefore, fasudil is an attractive antitumor drug candidate for the treatment of laryngeal carcinoma.
Collapse
Affiliation(s)
- Xiaowen Zhang
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Nan Wu
- The Core Laboratory for Public Health Science and Practice, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
35
|
Wada KI, Hosokawa K, Ito Y, Maeda M. Quantitative control of mitochondria transfer between live single cells using a microfluidic device. Biol Open 2017; 6:1960-1965. [PMID: 29092814 PMCID: PMC5769642 DOI: 10.1242/bio.024869] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Quantitative control of mitochondria transfer between live cells is a promising approach for genetic manipulation of mitochondrial DNA (mtDNA) because single mitochondrion transfer to a mtDNA-less (ρ0) cell potentially leads to homoplasmy of mtDNA. In this paper, we describe a method for quantitative control of mitochondria transfer between live single cells. For this purpose, we fabricated novel microfluidic devices having cell paring structures with a 4.1, 5.6 or 10.0 μm-length microtunnel. When cells were fused through a microtunnel using the Sendai virus envelope-based method, a strictured cytoplasmic connection was achieved with a length corresponding to that of the microtunnel. Elongation of the cytoplasmic connection led to a decrease in mitochondria transfer to the fusion partner. Moreover, some cell pairs that fused through a 10.0 μm-length microtunnel showed single mitochondrion transfer. Fused cells were spontaneously disconnected from each other when they were recovered in a normal culture medium. These results suggest that our cell fusion method can perform quantitative control of mitochondria transfer that includes a single mitochondrion transfer. Summary: We developed a novel mitochondria transfer platform using a microfluidic device, and succeeded in single mitochondrion transfer between live single cells.
Collapse
Affiliation(s)
- Ken-Ichi Wada
- Bioengineering Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Kazuo Hosokawa
- Bioengineering Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yoshihiro Ito
- Nano Medical Engineering Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Mizuo Maeda
- Bioengineering Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
36
|
Reichova A, Zatkova M, Bacova Z, Bakos J. Abnormalities in interactions of Rho GTPases with scaffolding proteins contribute to neurodevelopmental disorders. J Neurosci Res 2017; 96:781-788. [DOI: 10.1002/jnr.24200] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/09/2017] [Accepted: 10/30/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Alexandra Reichova
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences; Bratislava Slovakia
| | - Martina Zatkova
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences; Bratislava Slovakia
- Institute of Physiology; Comenius University, Faculty of Medicine; Bratislava Slovakia
| | - Zuzana Bacova
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences; Bratislava Slovakia
- Department of Normal and Pathological Physiology, Faculty of Medicine; Slovak Medical University; Bratislava Slovakia
| | - Jan Bakos
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences; Bratislava Slovakia
- Institute of Physiology; Comenius University, Faculty of Medicine; Bratislava Slovakia
| |
Collapse
|
37
|
Cartagena-Rivera AX, Van Itallie CM, Anderson JM, Chadwick RS. Apical surface supracellular mechanical properties in polarized epithelium using noninvasive acoustic force spectroscopy. Nat Commun 2017; 8:1030. [PMID: 29044161 PMCID: PMC5715111 DOI: 10.1038/s41467-017-01145-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 08/18/2017] [Indexed: 11/24/2022] Open
Abstract
Maintenance of epithelial tissue integrity requires coordination between cell–cell adherens junctions, tight junctions (TJ), and the perijunctional actomyosin cytoskeleton. Here we addressed the hypothesis that alterations in TJ structure and remodeling of the actomyosin cytoskeleton modify epithelial mechanics. Current methods to measure supracellular mechanical properties disrupt intact monolayers, therefore, we developed a novel method using noncontact acoustic frequency-modulation atomic force microscopy (FM-AFM) and tested it on MDCK polarized monolayers. Our results show that double knockdown (dKD) of ZO-1/ZO-2 elevates the apical epithelial tension and effective viscosity. Interestingly, epithelial tension is more sensitive to inhibition of myosin II ATPase activity than to inhibition of ROCK activity, but viscosity is highly sensitive to both. Additionally, we showed epithelial intercellular pulling forces at tricellular junctions and adhesion forces in dKD cells are elevated with an increase in contractility. In conclusion, FM-AFM enables the physiological and quantitative investigation of mechanics in intact epithelium. Determination of apical tension, fluidity, and intercellular adhesive forces in an epithelial monolayer are currently disruptive. Here the authors present a method using acoustic force microscopy to measure changes in these parameters upon tight junction structural alterations in a MDCK monolayer.
Collapse
Affiliation(s)
- Alexander X Cartagena-Rivera
- Section on Auditory Mechanics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Christina M Van Itallie
- Laboratory of Tight Junction Structure and Function, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - James M Anderson
- Laboratory of Tight Junction Structure and Function, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Richard S Chadwick
- Section on Auditory Mechanics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
38
|
Shapiro LP, Parsons RG, Koleske AJ, Gourley SL. Differential expression of cytoskeletal regulatory factors in the adolescent prefrontal cortex: Implications for cortical development. J Neurosci Res 2017; 95:1123-1143. [PMID: 27735056 PMCID: PMC5352542 DOI: 10.1002/jnr.23960] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 09/04/2016] [Accepted: 09/12/2016] [Indexed: 12/27/2022]
Abstract
The prevalence of depression, anxiety, schizophrenia, and drug and alcohol use disorders peaks during adolescence. Further, up to 50% of "adult" mental health disorders emerge in adolescence. During adolescence, the prefrontal cortex (PFC) undergoes dramatic structural reorganization, in which dendritic spines and synapses are refined, pruned, and stabilized. Understanding the molecular mechanisms that underlie these processes should help to identify factors that influence the development of psychiatric illness. Here we briefly discuss the anatomical connections of the medial and orbital prefrontal cortex (mPFC and OFC, respectively). We then present original findings suggesting that dendritic spines on deep-layer excitatory neurons in the mouse mPFC and OFC prune at different adolescent ages, with later pruning in the OFC. In parallel, we used Western blotting to define levels of several cytoskeletal regulatory proteins during early, mid-, and late adolescence, focusing on tropomyosin-related kinase receptor B (TrkB) and β1-integrin-containing receptors and select signaling partners. We identified regional differences in the levels of several proteins in early and midadolescence that then converged in early adulthood. We also observed age-related differences in TrkB levels, both full-length and truncated isoforms, Rho-kinase 2, and synaptophysin in both PFC subregions. Finally, we identified changes in protein levels in the dorsal and ventral hippocampus that were distinct from those in the PFC. We conclude with a general review of the manner in which TrkB- and β1-integrin-mediated signaling influences neuronal structure in the postnatal brain. Elucidating the role of cytoskeletal regulatory factors throughout adolescence may identify critical mechanisms of PFC development. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lauren P Shapiro
- Molecular and Systems Pharmacology, Emory University, Atlanta, Georgia
- Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
| | - Ryan G Parsons
- Department of Psychology and Neuroscience Institute, Graduate Program in Integrative Neuroscience, Program in Neuroscience, Stony Brook University, Stony Brook, New York
| | - Anthony J Koleske
- Department of Molecular Biophysics and Biochemistry, Department of Neurobiology, Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut
| | - Shannon L Gourley
- Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia
- Graduate Program in Neuroscience, Emory University, Atlanta, Georgia
| |
Collapse
|
39
|
Vasquez CG, Heissler SM, Billington N, Sellers JR, Martin AC. Drosophila non-muscle myosin II motor activity determines the rate of tissue folding. eLife 2016; 5. [PMID: 28035903 PMCID: PMC5201417 DOI: 10.7554/elife.20828] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 12/08/2016] [Indexed: 12/15/2022] Open
Abstract
Non-muscle cell contractility is critical for tissues to adopt shape changes. Although, the non-muscle myosin II holoenzyme (myosin) is a molecular motor that powers contraction of actin cytoskeleton networks, recent studies have questioned the importance of myosin motor activity cell and tissue shape changes. Here, combining the biochemical analysis of enzymatic and motile properties for purified myosin mutants with in vivo measurements of apical constriction for the same mutants, we show that in vivo constriction rate scales with myosin motor activity. We show that so-called phosphomimetic mutants of the Drosophila regulatory light chain (RLC) do not mimic the phosphorylated RLC state in vitro. The defect in the myosin motor activity in these mutants is evident in developing Drosophila embryos where tissue recoil following laser ablation is decreased compared to wild-type tissue. Overall, our data highlights that myosin activity is required for rapid cell contraction and tissue folding in developing Drosophila embryos. DOI:http://dx.doi.org/10.7554/eLife.20828.001
Collapse
Affiliation(s)
- Claudia G Vasquez
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Sarah M Heissler
- Laboratory of Molecular Physiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Neil Billington
- Laboratory of Molecular Physiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - James R Sellers
- Laboratory of Molecular Physiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Adam C Martin
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| |
Collapse
|
40
|
García-Rojo G, Fresno C, Vilches N, Díaz-Véliz G, Mora S, Aguayo F, Pacheco A, Parra-Fiedler N, Parra CS, Rojas PS, Tejos M, Aliaga E, Fiedler JL. The ROCK Inhibitor Fasudil Prevents Chronic Restraint Stress-Induced Depressive-Like Behaviors and Dendritic Spine Loss in Rat Hippocampus. Int J Neuropsychopharmacol 2016; 20:336-345. [PMID: 27927737 PMCID: PMC5409106 DOI: 10.1093/ijnp/pyw108] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 11/23/2016] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Dendritic arbor simplification and dendritic spine loss in the hippocampus, a limbic structure implicated in mood disorders, are assumed to contribute to symptoms of depression. These morphological changes imply modifications in dendritic cytoskeleton. Rho GTPases are regulators of actin dynamics through their effector Rho kinase. We have reported that chronic stress promotes depressive-like behaviors in rats along with dendritic spine loss in apical dendrites of hippocampal pyramidal neurons, changes associated with Rho kinase activation. The present study proposes that the Rho kinase inhibitor Fasudil may prevent the stress-induced behavior and dendritic spine loss. METHODS Adult male Sprague-Dawley rats were injected with saline or Fasudil (i.p., 10 mg/kg) starting 4 days prior to and maintained during the restraint stress procedure (2.5 h/d for 14 days). Nonstressed control animals were injected with saline or Fasudil for 18 days. At 24 hours after treatment, forced swimming test, Golgi-staining, and immuno-western blot were performed. RESULTS Fasudil prevented stress-induced immobility observed in the forced swimming test. On the other hand, Fasudil-treated control animals showed behavioral patterns similar to those of saline-treated controls. Furthermore, we observed that stress induced an increase in the phosphorylation of MYPT1 in the hippocampus, an exclusive target of Rho kinase. This change was accompanied by dendritic spine loss of apical dendrites of pyramidal hippocampal neurons. Interestingly, increased pMYPT1 levels and spine loss were both prevented by Fasudil administration. CONCLUSION Our findings suggest that Fasudil may prevent the development of abnormal behavior and spine loss induced by chronic stress by blocking Rho kinase activity.
Collapse
Affiliation(s)
- Gonzalo García-Rojo
- Laboratory of Neuroplasticity and Neurogenetics, Department of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile (Mr García-Rojo, Ms Vilches, Mr Aguayo, Ms Pacheco, Mr Parra-Fiedler, Mr Parra, Dr Rojas, Ms Tejos, and Dr Fiedler); CONICET, Universidad Católica de Córdoba, Córdoba, Argentina (Dr Fresno); Laboratorio Farmacología del Comportamiento, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile (Ms Díaz-Véliz and Mr Mora); Faculty of Medicine, School of Pharmacy, Universidad Andres Bello, Santiago, Chile (Dr Rojas, present address); Department of Kinesiology, Faculty of Health Sciences, Universidad Católica del Maule, Talca, Chile (Dr Aliaga)
| | - Cristóbal Fresno
- Laboratory of Neuroplasticity and Neurogenetics, Department of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile (Mr García-Rojo, Ms Vilches, Mr Aguayo, Ms Pacheco, Mr Parra-Fiedler, Mr Parra, Dr Rojas, Ms Tejos, and Dr Fiedler); CONICET, Universidad Católica de Córdoba, Córdoba, Argentina (Dr Fresno); Laboratorio Farmacología del Comportamiento, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile (Ms Díaz-Véliz and Mr Mora); Faculty of Medicine, School of Pharmacy, Universidad Andres Bello, Santiago, Chile (Dr Rojas, present address); Department of Kinesiology, Faculty of Health Sciences, Universidad Católica del Maule, Talca, Chile (Dr Aliaga)
| | - Natalia Vilches
- Laboratory of Neuroplasticity and Neurogenetics, Department of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile (Mr García-Rojo, Ms Vilches, Mr Aguayo, Ms Pacheco, Mr Parra-Fiedler, Mr Parra, Dr Rojas, Ms Tejos, and Dr Fiedler); CONICET, Universidad Católica de Córdoba, Córdoba, Argentina (Dr Fresno); Laboratorio Farmacología del Comportamiento, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile (Ms Díaz-Véliz and Mr Mora); Faculty of Medicine, School of Pharmacy, Universidad Andres Bello, Santiago, Chile (Dr Rojas, present address); Department of Kinesiology, Faculty of Health Sciences, Universidad Católica del Maule, Talca, Chile (Dr Aliaga)
| | - Gabriela Díaz-Véliz
- Laboratory of Neuroplasticity and Neurogenetics, Department of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile (Mr García-Rojo, Ms Vilches, Mr Aguayo, Ms Pacheco, Mr Parra-Fiedler, Mr Parra, Dr Rojas, Ms Tejos, and Dr Fiedler); CONICET, Universidad Católica de Córdoba, Córdoba, Argentina (Dr Fresno); Laboratorio Farmacología del Comportamiento, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile (Ms Díaz-Véliz and Mr Mora); Faculty of Medicine, School of Pharmacy, Universidad Andres Bello, Santiago, Chile (Dr Rojas, present address); Department of Kinesiology, Faculty of Health Sciences, Universidad Católica del Maule, Talca, Chile (Dr Aliaga)
| | - Sergio Mora
- Laboratory of Neuroplasticity and Neurogenetics, Department of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile (Mr García-Rojo, Ms Vilches, Mr Aguayo, Ms Pacheco, Mr Parra-Fiedler, Mr Parra, Dr Rojas, Ms Tejos, and Dr Fiedler); CONICET, Universidad Católica de Córdoba, Córdoba, Argentina (Dr Fresno); Laboratorio Farmacología del Comportamiento, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile (Ms Díaz-Véliz and Mr Mora); Faculty of Medicine, School of Pharmacy, Universidad Andres Bello, Santiago, Chile (Dr Rojas, present address); Department of Kinesiology, Faculty of Health Sciences, Universidad Católica del Maule, Talca, Chile (Dr Aliaga)
| | - Felipe Aguayo
- Laboratory of Neuroplasticity and Neurogenetics, Department of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile (Mr García-Rojo, Ms Vilches, Mr Aguayo, Ms Pacheco, Mr Parra-Fiedler, Mr Parra, Dr Rojas, Ms Tejos, and Dr Fiedler); CONICET, Universidad Católica de Córdoba, Córdoba, Argentina (Dr Fresno); Laboratorio Farmacología del Comportamiento, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile (Ms Díaz-Véliz and Mr Mora); Faculty of Medicine, School of Pharmacy, Universidad Andres Bello, Santiago, Chile (Dr Rojas, present address); Department of Kinesiology, Faculty of Health Sciences, Universidad Católica del Maule, Talca, Chile (Dr Aliaga)
| | - Aníbal Pacheco
- Laboratory of Neuroplasticity and Neurogenetics, Department of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile (Mr García-Rojo, Ms Vilches, Mr Aguayo, Ms Pacheco, Mr Parra-Fiedler, Mr Parra, Dr Rojas, Ms Tejos, and Dr Fiedler); CONICET, Universidad Católica de Córdoba, Córdoba, Argentina (Dr Fresno); Laboratorio Farmacología del Comportamiento, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile (Ms Díaz-Véliz and Mr Mora); Faculty of Medicine, School of Pharmacy, Universidad Andres Bello, Santiago, Chile (Dr Rojas, present address); Department of Kinesiology, Faculty of Health Sciences, Universidad Católica del Maule, Talca, Chile (Dr Aliaga)
| | - Nicolás Parra-Fiedler
- Laboratory of Neuroplasticity and Neurogenetics, Department of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile (Mr García-Rojo, Ms Vilches, Mr Aguayo, Ms Pacheco, Mr Parra-Fiedler, Mr Parra, Dr Rojas, Ms Tejos, and Dr Fiedler); CONICET, Universidad Católica de Córdoba, Córdoba, Argentina (Dr Fresno); Laboratorio Farmacología del Comportamiento, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile (Ms Díaz-Véliz and Mr Mora); Faculty of Medicine, School of Pharmacy, Universidad Andres Bello, Santiago, Chile (Dr Rojas, present address); Department of Kinesiology, Faculty of Health Sciences, Universidad Católica del Maule, Talca, Chile (Dr Aliaga)
| | - Claudio S. Parra
- Laboratory of Neuroplasticity and Neurogenetics, Department of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile (Mr García-Rojo, Ms Vilches, Mr Aguayo, Ms Pacheco, Mr Parra-Fiedler, Mr Parra, Dr Rojas, Ms Tejos, and Dr Fiedler); CONICET, Universidad Católica de Córdoba, Córdoba, Argentina (Dr Fresno); Laboratorio Farmacología del Comportamiento, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile (Ms Díaz-Véliz and Mr Mora); Faculty of Medicine, School of Pharmacy, Universidad Andres Bello, Santiago, Chile (Dr Rojas, present address); Department of Kinesiology, Faculty of Health Sciences, Universidad Católica del Maule, Talca, Chile (Dr Aliaga)
| | - Paulina S. Rojas
- Laboratory of Neuroplasticity and Neurogenetics, Department of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile (Mr García-Rojo, Ms Vilches, Mr Aguayo, Ms Pacheco, Mr Parra-Fiedler, Mr Parra, Dr Rojas, Ms Tejos, and Dr Fiedler); CONICET, Universidad Católica de Córdoba, Córdoba, Argentina (Dr Fresno); Laboratorio Farmacología del Comportamiento, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile (Ms Díaz-Véliz and Mr Mora); Faculty of Medicine, School of Pharmacy, Universidad Andres Bello, Santiago, Chile (Dr Rojas, present address); Department of Kinesiology, Faculty of Health Sciences, Universidad Católica del Maule, Talca, Chile (Dr Aliaga)
| | - Macarena Tejos
- Laboratory of Neuroplasticity and Neurogenetics, Department of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile (Mr García-Rojo, Ms Vilches, Mr Aguayo, Ms Pacheco, Mr Parra-Fiedler, Mr Parra, Dr Rojas, Ms Tejos, and Dr Fiedler); CONICET, Universidad Católica de Córdoba, Córdoba, Argentina (Dr Fresno); Laboratorio Farmacología del Comportamiento, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile (Ms Díaz-Véliz and Mr Mora); Faculty of Medicine, School of Pharmacy, Universidad Andres Bello, Santiago, Chile (Dr Rojas, present address); Department of Kinesiology, Faculty of Health Sciences, Universidad Católica del Maule, Talca, Chile (Dr Aliaga)
| | - Esteban Aliaga
- Laboratory of Neuroplasticity and Neurogenetics, Department of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile (Mr García-Rojo, Ms Vilches, Mr Aguayo, Ms Pacheco, Mr Parra-Fiedler, Mr Parra, Dr Rojas, Ms Tejos, and Dr Fiedler); CONICET, Universidad Católica de Córdoba, Córdoba, Argentina (Dr Fresno); Laboratorio Farmacología del Comportamiento, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile (Ms Díaz-Véliz and Mr Mora); Faculty of Medicine, School of Pharmacy, Universidad Andres Bello, Santiago, Chile (Dr Rojas, present address); Department of Kinesiology, Faculty of Health Sciences, Universidad Católica del Maule, Talca, Chile (Dr Aliaga)
| | - Jenny L. Fiedler
- Laboratory of Neuroplasticity and Neurogenetics, Department of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile (Mr García-Rojo, Ms Vilches, Mr Aguayo, Ms Pacheco, Mr Parra-Fiedler, Mr Parra, Dr Rojas, Ms Tejos, and Dr Fiedler); CONICET, Universidad Católica de Córdoba, Córdoba, Argentina (Dr Fresno); Laboratorio Farmacología del Comportamiento, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile (Ms Díaz-Véliz and Mr Mora); Faculty of Medicine, School of Pharmacy, Universidad Andres Bello, Santiago, Chile (Dr Rojas, present address); Department of Kinesiology, Faculty of Health Sciences, Universidad Católica del Maule, Talca, Chile (Dr Aliaga)
| |
Collapse
|
41
|
Matas-Rico E, van Veen M, Leyton-Puig D, van den Berg J, Koster J, Kedziora KM, Molenaar B, Weerts MJA, de Rink I, Medema RH, Giepmans BNG, Perrakis A, Jalink K, Versteeg R, Moolenaar WH. Glycerophosphodiesterase GDE2 Promotes Neuroblastoma Differentiation through Glypican Release and Is a Marker of Clinical Outcome. Cancer Cell 2016; 30:548-562. [PMID: 27693046 DOI: 10.1016/j.ccell.2016.08.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 06/06/2016] [Accepted: 08/26/2016] [Indexed: 02/06/2023]
Abstract
Neuroblastoma is a pediatric embryonal malignancy characterized by impaired neuronal differentiation. A better understanding of neuroblastoma differentiation is essential for developing new therapeutic approaches. GDE2 (encoded by GDPD5) is a six-transmembrane-domain glycerophosphodiesterase that promotes embryonic neurogenesis. We find that high GDPD5 expression is strongly associated with favorable outcome in neuroblastoma. GDE2 induces differentiation of neuroblastoma cells, suppresses cell motility, and opposes RhoA-driven neurite retraction. GDE2 alters the Rac-RhoA activity balance and the expression of multiple differentiation-associated genes. Mechanistically, GDE2 acts by cleaving (in cis) and releasing glycosylphosphatidylinositol-anchored glypican-6, a putative co-receptor. A single point mutation in the ectodomain abolishes GDE2 function. Our results reveal GDE2 as a cell-autonomous inducer of neuroblastoma differentiation with prognostic significance and potential therapeutic value.
Collapse
Affiliation(s)
- Elisa Matas-Rico
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Michiel van Veen
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Daniela Leyton-Puig
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Jeroen van den Berg
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Jan Koster
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Katarzyna M Kedziora
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Bas Molenaar
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Marjolein J A Weerts
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Iris de Rink
- Deep Sequencing Core Facility, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - René H Medema
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Ben N G Giepmans
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Anastassis Perrakis
- Division of Biochemistry, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Kees Jalink
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Rogier Versteeg
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Wouter H Moolenaar
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands.
| |
Collapse
|
42
|
Xiang X, Li S, Zhuang X, Shi L. Arhgef1 negatively regulates neurite outgrowth through activation of RhoA signaling pathways. FEBS Lett 2016; 590:2940-55. [PMID: 27489999 DOI: 10.1002/1873-3468.12339] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 06/20/2016] [Accepted: 07/25/2016] [Indexed: 11/06/2022]
Abstract
Neurite outgrowth is essential for the establishment of functional neuronal connections during brain development. This study identifies that Arhgef1 is predominantly expressed in early neuronal developmental stages and negatively regulates neurite outgrowth. Knockdown of Arhgef1 in either Neuro-2a cells or primary cortical neurons leads to excess growth of neurites, whereas overexpression of Arhgef1 prominently restricts neurite formation. Arhgef1 strongly activates RhoA activity while concomitantly inhibits Rac1 and Cdc42 activities. Pharmacological blockade of RhoA activity restores normal neurite outgrowth in Arhgef1-overexpressed neurons. Importantly, Arhgef1 promotes F-actin polymerization in neurons, probably through inhibiting the activity of the actin-depolymerizing factor cofilin. Collectively, these findings reveal that Arhgef1 functions as a negative regulator of neurite outgrowth through regulating RhoA-cofilin pathway and actin dynamics.
Collapse
Affiliation(s)
- Xiaoliang Xiang
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, Jinan University, Guangzhou, Guangdong, China
| | - Shengnan Li
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, Jinan University, Guangzhou, Guangdong, China
| | - Xiaoji Zhuang
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, Jinan University, Guangzhou, Guangdong, China
| | - Lei Shi
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
43
|
Jia XF, Ye F, Wang YB, Feng DX. ROCK inhibition enhances neurite outgrowth in neural stem cells by upregulating YAP expression in vitro. Neural Regen Res 2016; 11:983-7. [PMID: 27482229 PMCID: PMC4962598 DOI: 10.4103/1673-5374.184499] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Spontaneous axonal regeneration of neurons does not occur after spinal cord injury because of inhibition by myelin and other inhibitory factors. Studies have demonstrated that blocking the Rho/Rho-kinase (ROCK) pathway can promote neurite outgrowth in spinal cord injury models. In the present study, we investigated neurite outgrowth and neuronal differentiation in neural stem cells from the mouse subventricular zone after inhibition of ROCK in vitro. Inhibition of ROCK with Y-27632 increased neurite length, enhanced neuronal differentiation, and upregulated the expression of two major signaling pathway effectors, phospho-Akt and phospho-mitogen-activated protein kinase, and the Hippo pathway effector YAP. These results suggest that inhibition of ROCK mediates neurite outgrowth in neural stem cells by activating the Hippo signaling pathway.
Collapse
Affiliation(s)
- Xu-Feng Jia
- Department of Orthopedic Surgery, Jianyang People's Hospital of Sichuan Province, Jianyang, Sichuan Province, China
| | - Fei Ye
- Department of Spine Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yan-Bo Wang
- Department of Spine Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Da-Xiong Feng
- Department of Spine Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| |
Collapse
|
44
|
Fusco L, Lefort R, Smith K, Benmansour F, Gonzalez G, Barillari C, Rinn B, Fleuret F, Fua P, Pertz O. Computer vision profiling of neurite outgrowth dynamics reveals spatiotemporal modularity of Rho GTPase signaling. J Cell Biol 2016; 212:91-111. [PMID: 26728857 PMCID: PMC4700477 DOI: 10.1083/jcb.201506018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
NeuriteTracker is a computer vision approach used to analyze neuronal morphodynamics and to examine spatiotemporal Rho GTPase signaling networks regulating neurite outgrowth. Rho guanosine triphosphatases (GTPases) control the cytoskeletal dynamics that power neurite outgrowth. This process consists of dynamic neurite initiation, elongation, retraction, and branching cycles that are likely to be regulated by specific spatiotemporal signaling networks, which cannot be resolved with static, steady-state assays. We present NeuriteTracker, a computer-vision approach to automatically segment and track neuronal morphodynamics in time-lapse datasets. Feature extraction then quantifies dynamic neurite outgrowth phenotypes. We identify a set of stereotypic neurite outgrowth morphodynamic behaviors in a cultured neuronal cell system. Systematic RNA interference perturbation of a Rho GTPase interactome consisting of 219 proteins reveals a limited set of morphodynamic phenotypes. As proof of concept, we show that loss of function of two distinct RhoA-specific GTPase-activating proteins (GAPs) leads to opposite neurite outgrowth phenotypes. Imaging of RhoA activation dynamics indicates that both GAPs regulate different spatiotemporal Rho GTPase pools, with distinct functions. Our results provide a starting point to dissect spatiotemporal Rho GTPase signaling networks that regulate neurite outgrowth.
Collapse
Affiliation(s)
- Ludovico Fusco
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Riwal Lefort
- Institut Dalla Molle d'Intelligence Artificielle Perceptive (IDIAP Research Institute), 1920 Martigny, Switzerland
| | - Kevin Smith
- Computer Vision Laboratory, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Fethallah Benmansour
- Computer Vision Laboratory, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - German Gonzalez
- Computer Vision Laboratory, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Caterina Barillari
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule, 4058 Basel, Switzerland
| | - Bernd Rinn
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule, 4058 Basel, Switzerland
| | - Francois Fleuret
- Institut Dalla Molle d'Intelligence Artificielle Perceptive (IDIAP Research Institute), 1920 Martigny, Switzerland
| | - Pascal Fua
- Computer Vision Laboratory, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Olivier Pertz
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| |
Collapse
|
45
|
Yu JZ, Chen C, Zhang Q, Zhao YF, Feng L, Zhang HF, Meng J, Ma CG, Xiao BG. Changes of synapses in experimental autoimmune encephalomyelitis by using Fasudil. Wound Repair Regen 2016; 24:317-27. [PMID: 26789651 DOI: 10.1111/wrr.12407] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 12/22/2015] [Indexed: 11/29/2022]
Abstract
The ROCK signaling pathway is involved in numerous fundamental cellular functions such as cell migration, apoptosis, inflammatory responses, and neurite outgrowth. Previous studies demonstrate that Fasudil exhibited therapeutic potential of experimental autoimmune encephalomyelitis (EAE) possibly through immune-modulation and anti-inflammation. In this study, we observed the effect of Fasudil on synaptic protection of EAE mice. Fasudil ameliorated the clinical severity of EAE and inhibited Rho kinase (ROCK), especially ROCK II, in brain and spinal cord of EAE mice. Protein extracts from spinal cord of Fasudil-treated EAE mice promoted the formation of neurite outgrowth when co-cultured with primary neurons, indicating that peripheral administration of Fasudil can enter the central nervous system (CNS) and exhibited its biological effect on the formation of neurite outgrowth. Synapse-related molecule synaptophysin was enhanced, and CRMP-2, AMPA receptor, and GSK-3β were declined in spinal cord of Fasudil-treated mice. Neurotrophic factor BDNF and GDNF as well as immunomodulatory cytokine IL-10 in spinal cord were elevated in Fasudil-treated mice, while inflammatory cytokine IL-17, IL-1β, IL-6, and TNF-α were obviously inhibited, accompanied by the decrease of inflammatory M1 iNOS and the increase of anti-inflammatory M2 Arg-1, providing a microenvironment that contributes to synaptic protection. Our results indicate that Fasudil treatment protected against synaptic damage and promoted synaptic formation, which may be related with increased neurotrophic factors as well as decreased inflammatory microenvironment in the CNS of EAE mice.
Collapse
Affiliation(s)
- Jie-Zhong Yu
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China
| | - Chan Chen
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Qiong Zhang
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Yong-Fei Zhao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Ling Feng
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China
| | - Hai-Fei Zhang
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China
| | - Jian Meng
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China
| | - Cun-Gen Ma
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong, China.,"2011" Collaborative Innovation Center/Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Bao-Guo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| |
Collapse
|
46
|
Antoine-Bertrand J, Fu M, Lamarche-Vane N. Direct measurement of oscillatory RhoA activity in embryonic cortical neurons stimulated with the axon guidance cue netrin-1 using fluorescence resonance energy transfer. Biol Cell 2016; 108:115-26. [PMID: 26787017 DOI: 10.1111/boc.201500077] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 01/15/2016] [Indexed: 12/27/2022]
Abstract
BACKGROUND INFORMATION Rho GTPases play an essential role during the development of the nervous system. They induce cytoskeletal rearrangements that are critical for the regulation of axon outgrowth and guidance. It is generally accepted that Rac1 and Cdc42 are positive regulators of axon outgrowth and guidance, whereas RhoA is a negative regulator. However, spatiotemporal control of their activity can modify the function of Rho GTPases during axonal morphogenesis. Signalling downstream of the axon guidance cue netrin-1 and its receptor deleted in colorectal cancer (DCC) triggers the activation of Rac1 and the inhibition of RhoA to promote axon outgrowth. However, our previous work also suggests that netrin-1/DCC signalling can activate RhoA in a time- and region-specific manner. RESULTS Here, we visualised RhoA activation in response to netrin-1 in live embryonic cortical neurons using fluorescence resonance energy transfer. RhoA activity oscillated in unstimulated neurons and netrin-1 increased the amplitude of the oscillations in growth cones after 5 min of stimulation. Within this period of time, netrin-1 transiently increased RhoA activity and modulated the pattern of RhoA oscillations. We found that the timing of netrin-1-induced RhoA activation was different in whole neurons, cell bodies and growth cones. CONCLUSIONS We conclude that netrin-1 modulates the spatiotemporal activation of RhoA in embryonic cortical neurons. SIGNIFICANCE This study demonstrates for the first time the short-term localised activation of RhoA in neuronal growth cones by the axon guidance cue netrin-1.
Collapse
Affiliation(s)
- Judith Antoine-Bertrand
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada.,Cancer Research Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada
| | - Min Fu
- Cancer Research Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada
| | - Nathalie Lamarche-Vane
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada.,Cancer Research Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada
| |
Collapse
|
47
|
Cartocci V, Segatto M, Di Tunno I, Leone S, Pfrieger FW, Pallottini V. Modulation of the Isoprenoid/Cholesterol Biosynthetic Pathway During Neuronal Differentiation In Vitro. J Cell Biochem 2016; 117:2036-44. [PMID: 27392312 DOI: 10.1002/jcb.25500] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 01/25/2016] [Indexed: 12/27/2022]
Abstract
During differentiation, neurons acquire their typical shape and functional properties. At present, it is unclear, whether this important developmental step involves metabolic changes. Here, we studied the contribution of the mevalonate (MVA) pathway to neuronal differentiation using the mouse neuroblastoma cell line N1E-115 as experimental model. Our results show that during differentiation, the activity of 3-hydroxy 3-methylglutaryl Coenzyme A reductase (HMGR), a key enzyme of MVA pathway, and the level of Low Density Lipoprotein receptor (LDLr) decrease, whereas the level of LDLr-related protein-1 (LRP1) and the dimerization of Scavanger Receptor B1 (SRB-1) rise. Pharmacologic inhibition of HMGR by simvastatin accelerated neuronal differentiation by modulating geranylated proteins. Collectively, our data suggest that during neuronal differentiation, the activity of the MVA pathway decreases and we postulate that any interference with this process impacts neuronal morphology and function. Therefore, the MVA pathway appears as an attractive pharmacological target to modulate neurological and metabolic symptoms of developmental neuropathologies. J. Cell. Biochem. 117: 2036-2044, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Veronica Cartocci
- Department of Science, Biomedical and Technology Science Section, University Roma Tre, Viale Marconi, 446, 00146, Rome, Italy
| | - Marco Segatto
- Department of Biosciences, University of Milan, Via Giovanni Celoria, 26, 20133, Milan, Italy
| | - Ilenia Di Tunno
- Department of Science, Biomedical and Technology Science Section, University Roma Tre, Viale Marconi, 446, 00146, Rome, Italy
| | - Stefano Leone
- Department of Science, Biomedical and Technology Science Section, University Roma Tre, Viale Marconi, 446, 00146, Rome, Italy
| | - Frank W Pfrieger
- Institute of Cellular and Integrative Neurosciences (INCI) CNRS UPR 3212, University of Strasbourg, 5 rue Blaise Pascal, 67084, Strasbourg, France
| | - Valentina Pallottini
- Department of Science, Biomedical and Technology Science Section, University Roma Tre, Viale Marconi, 446, 00146, Rome, Italy
| |
Collapse
|
48
|
Kamishibahara Y, Kawaguchi H, Shimizu N. Rho kinase inhibitor Y-27632 promotes neuronal differentiation in mouse embryonic stem cells via phosphatidylinositol 3-kinase. Neurosci Lett 2016; 615:44-9. [PMID: 26797580 DOI: 10.1016/j.neulet.2016.01.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 01/14/2016] [Accepted: 01/14/2016] [Indexed: 10/22/2022]
Abstract
Rho kinase (ROCK) regulates the functions of several target proteins via its kinase activity. Therefore, ROCK activity inhibition may provide new possibilities of controlling the in vitro neuronal differentiation of embryonic stem (ES) cells. When we investigated the effects of the ROCK inhibitor Y-27632 on ES cell differentiation, we found that this inhibitor promoted the differentiation of these cells into neurons. Furthermore, we found that ROCK inhibition may promote the neuronal differentiation of ES cells by activating extracellular signal-regulated kinase (ERK) involved in the ERK signaling pathway. In this study, we investigated the effects of specific inhibitors of several cellular signaling components on the promotion of neuronal differentiation in ES cells to clarify the roles of cellular signaling pathways in the ROCK inhibitor-mediated cell differentiation process. Our results suggest that ERK may be activated via the Ras/Raf/MEK, the PI3K/PKC, or the Cdc42/Rac signaling pathways in the ROCK inhibitor-mediated promotion of neuronal differentiation in ES cells.
Collapse
Affiliation(s)
- Yu Kamishibahara
- Graduate School of Life Sciences, Toyo University, 1-1-1 Izumino, Itakura-machi, Ora-gun, Gunma 374-0193, Japan.
| | - Hideo Kawaguchi
- Graduate School of Life Sciences, Toyo University, 1-1-1 Izumino, Itakura-machi, Ora-gun, Gunma 374-0193, Japan.
| | - Norio Shimizu
- Graduate School of Life Sciences, Toyo University, 1-1-1 Izumino, Itakura-machi, Ora-gun, Gunma 374-0193, Japan; Bio-Nano Electronics Research Center, Toyo University, 2100 Kujirai, Kawagoe-shi, Saitama 350-8585, Japan.
| |
Collapse
|
49
|
Ishihara S, Yasuda M, Ishizu A, Ishikawa M, Shirato H, Haga H. Activating transcription factor 5 enhances radioresistance and malignancy in cancer cells. Oncotarget 2016; 6:4602-14. [PMID: 25682872 PMCID: PMC4467102 DOI: 10.18632/oncotarget.2912] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 12/11/2014] [Indexed: 11/25/2022] Open
Abstract
Radiotherapy is effective for treating various types of tumors. However, some cancer cells survive after irradiation and repopulate tumors with highly malignant phenotypes that correlate with poor prognosis. It is not known how cancer cells survive and generate malignant tumors after irradiation. Here, we show that activating transcription factor 5 (ATF5) promotes radioresistance and malignancy in cancer cells after irradiation. In the G1-S phase of the cell cycle, cancer cells express high levels of ATF5, which promotes cell cycle progression and thereby increases radioresistance. Furthermore, ATF5 increases malignant phenotypes, such as cell growth and invasiveness, in cancer cells in vitro and in vivo. We have identified a new mechanism for the regeneration of highly malignant tumors after irradiation and shown that ATF5 plays a key role in the process.
Collapse
Affiliation(s)
- Seiichiro Ishihara
- Faculty of Advanced Life Science, Hokkaido University, Kita-ku, Sapporo 060-0810, Japan.,Research Center for Cooperative Projects, Graduate School of Medicine, Hokkaido University, Kita-ku, Sapporo 060-8638, Japan
| | - Motoaki Yasuda
- Department of Oral Pathobiological Science, Graduate School of Dental Medicine, Hokkaido University, Kita-ku, Sapporo 060-8586, Japan
| | - Akihiro Ishizu
- Division of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan
| | - Masayori Ishikawa
- Department of Medical Physics, Graduate School of Medicine, Hokkaido University, Kita-ku, Sapporo 060-8638, Japan
| | - Hiroki Shirato
- Department of Radiology, Graduate School of Medicine, Hokkaido University, Kita-ku, Sapporo 060-8638, Japan
| | - Hisashi Haga
- Faculty of Advanced Life Science, Hokkaido University, Kita-ku, Sapporo 060-0810, Japan.,Research Center for Cooperative Projects, Graduate School of Medicine, Hokkaido University, Kita-ku, Sapporo 060-8638, Japan
| |
Collapse
|
50
|
Sari-Hassoun M, Clement MJ, Hamdi I, Bollot G, Bauvais C, Joshi V, Toma F, Burgo A, Cailleret M, Rosales-Hernández MC, Macias Pérez ME, Chabane-Sari D, Curmi PA. Cucurbitacin I elicits the formation of actin/phospho-myosin II co-aggregates by stimulation of the RhoA/ROCK pathway and inhibition of LIM-kinase. Biochem Pharmacol 2015; 102:45-63. [PMID: 26707799 DOI: 10.1016/j.bcp.2015.12.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 12/15/2015] [Indexed: 12/11/2022]
Abstract
Cucurbitacins are cytotoxic triterpenoid sterols isolated from plants. One of their earliest cellular effect is the aggregation of actin associated with blockage of cell migration and division that eventually lead to apoptosis. We unravel here that cucurbitacin I actually induces the co-aggregation of actin with phospho-myosin II. This co-aggregation most probably results from the stimulation of the Rho/ROCK pathway and the direct inhibition of the LIMKinase. We further provide data that suggest that the formation of these co-aggregates is independent of a putative pro-oxidant status of cucurbitacin I. The results help to understand the impact of cucurbitacins on signal transduction and actin dynamics and open novel perspectives to use it as drug candidates for cancer research.
Collapse
Affiliation(s)
- Meryem Sari-Hassoun
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France; Laboratoire des Produits Naturels, LAPRONA, Université Abou Bekr Belkaid, Tlemcen 13000, Algeria
| | - Marie-Jeanne Clement
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France
| | - Imane Hamdi
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France
| | | | | | - Vandana Joshi
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France
| | - Flavio Toma
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France
| | - Andrea Burgo
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France
| | - Michel Cailleret
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France
| | - Martha Cecilia Rosales-Hernández
- Laboratorio de Biofísica y Biocatalisis, Sección de Estudios de Posgrado e Investigación de la Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, México D.F. 11340, México
| | - Martha Edith Macias Pérez
- Laboratorio de Biofísica y Biocatalisis, Sección de Estudios de Posgrado e Investigación de la Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, México D.F. 11340, México
| | - Daoudi Chabane-Sari
- Laboratoire des Produits Naturels, LAPRONA, Université Abou Bekr Belkaid, Tlemcen 13000, Algeria
| | - Patrick A Curmi
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France.
| |
Collapse
|