1
|
Scarisbrick IA. PARting Neuroinflammation with Protease-Activated Receptor 2 Pepducins. J Pharmacol Exp Ther 2024; 388:8-11. [PMID: 38169447 DOI: 10.1124/jpet.123.001893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/26/2023] [Indexed: 01/05/2024] Open
Affiliation(s)
- Isobel A Scarisbrick
- Center for Regenerative Biotherapeutics, Department of Physical Medicine and Rehabilitation, and Department of Physiology and Biomedical Engineering, Mayo Clinic Graduate School of Biomedical Sciences, Regenerative Sciences, Rochester, Minnesota
| |
Collapse
|
2
|
Soeda M, Ohka S, Nishizawa D, Iseki M, Yamaguchi K, Arita H, Hanaoka K, Kato J, Ogawa S, Hiranuma A, Hasegawa J, Nakayama K, Ebata Y, Hayashida M, Ichinohe T, Fukuda KI, Ikeda K. Single-Nucleotide Polymorphisms of the PAR2 and IL-17A Genes Are Significantly Associated with Chronic Pain. Int J Mol Sci 2023; 24:17627. [PMID: 38139455 PMCID: PMC10744199 DOI: 10.3390/ijms242417627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Patients with chronic pain are affected psychologically and socially. There are also individual differences in treatment efficacy. Insufficient research has been conducted on genetic polymorphisms that are related to individual differences in the susceptibility to chronic pain. Autoimmune disorders can lead to inflammation and chronic pain; therefore, we focused on the autoimmune-related protease-activated receptor 2 (PAR2/F2RL1) and interleukin 17A (IL-17A/IL17A) genes. PAR2 and IL-17A are associated with autoimmune diseases that lead to chronic pain, and PAR2 regulates T-helper (Th) cell activation and differentiation. We hypothesized that the PAR2 and IL-17A genes are associated with chronic pain. The present study used a case-control design to statistically examine associations between genetic polymorphisms and the vulnerability to chronic pain. The rs2243057 polymorphism of the PAR2 gene and rs3819025 polymorphism of the IL-17A gene were previously reported to be associated with pain- or autoimmune-related phenotypes. Thus, these polymorphisms were investigated in the present study. We found that both rs2243057 and rs3819025 were significantly associated with a susceptibility to chronic pain. The present findings revealed autoimmune-related genetic factors that are involved in individual differences in chronic pain, further aiding understanding of the pathomechanism that underlies chronic pain and possibly contributing to future personalized medicine.
Collapse
Affiliation(s)
- Moe Soeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (M.S.); (S.O.); (D.N.)
- Department of Oral Health and Clinical Science, Tokyo Dental College, Tokyo 101-0061, Japan
| | - Seii Ohka
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (M.S.); (S.O.); (D.N.)
| | - Daisuke Nishizawa
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (M.S.); (S.O.); (D.N.)
| | - Masako Iseki
- Department of Anesthesiology & Pain Medicine, Juntendo University School of Medicine, Tokyo 113-8431, Japan; (M.I.)
| | - Keisuke Yamaguchi
- Department of Anesthesiology & Pain Medicine, Juntendo University School of Medicine, Tokyo 113-8431, Japan; (M.I.)
| | - Hideko Arita
- Department of Anesthesiology, Pain Relief Center, JR Tokyo General Hospital, Tokyo 151-8528, Japan; (H.A.); (K.H.)
| | - Kazuo Hanaoka
- Department of Anesthesiology, Pain Relief Center, JR Tokyo General Hospital, Tokyo 151-8528, Japan; (H.A.); (K.H.)
| | - Jitsu Kato
- Department of Anesthesiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Setsuro Ogawa
- University Research Center, Nihon University, Tokyo 173-8610, Japan
| | - Ayako Hiranuma
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (M.S.); (S.O.); (D.N.)
- Department of Surgery, Toho University Sakura Medical Center, Chiba 285-8741, Japan
| | - Junko Hasegawa
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (M.S.); (S.O.); (D.N.)
| | - Kyoko Nakayama
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (M.S.); (S.O.); (D.N.)
| | - Yuko Ebata
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (M.S.); (S.O.); (D.N.)
| | - Masakazu Hayashida
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (M.S.); (S.O.); (D.N.)
- Department of Anesthesiology & Pain Medicine, Juntendo University School of Medicine, Tokyo 113-8431, Japan; (M.I.)
- Department of Anesthesiology, Saitama Medical University International Medical Center, Saitama 350-1298, Japan
| | - Tatsuya Ichinohe
- Department of Dental Anesthesiology, Tokyo Dental College, Tokyo 101-0061, Japan;
| | - Ken-ichi Fukuda
- Department of Oral Health and Clinical Science, Tokyo Dental College, Tokyo 101-0061, Japan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (M.S.); (S.O.); (D.N.)
| |
Collapse
|
3
|
Bian Z, Liu X, Feng T, Yu H, Hu X, Hu X, Bian Y, Sun H, Tadokoro K, Takemoto M, Yunoki T, Nakano Y, Fukui Y, Morihara R, Abe K, Yamashita T. Protective Effect of Rivaroxaban Against Amyloid Pathology and Neuroinflammation Through Inhibiting PAR-1 and PAR-2 in Alzheimer's Disease Mice. J Alzheimers Dis 2022; 86:111-123. [PMID: 35001892 DOI: 10.3233/jad-215318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Recent studies have revealed that atrial fibrillation (AF) patients have a high risk of developing cognitive impairment, vascular dementia, and Alzheimer's disease (AD). Some reports suggest that the application of oral anticoagulant with an appropriate dose may have a preventive effect on AD. However, which oral anticoagulant drug is more appropriate for preventing AD and the underlying mechanism(s) is still unknown. OBJECTIVE The aim of the present study was to assess the treatment effect of rivaroxaban administration as well as investigate the roles of PAR-1 and PAR-2 in the AD + CAA mice model. METHODS In the present study, we compared a traditional oral anticoagulant, warfarin, and a direct oral anticoagulant (DOAC), rivaroxaban, via long-term administration to an AD with cerebral amyloid angiopathy (CAA) mice model. RESULTS Rivaroxaban treatment attenuated neuroinflammation, blood-brain barrier dysfunction, memory deficits, and amyloid-β deposition through PAR-1/PAR-2 inhibition in the AD + CAA mice model compared with warfarin and no-treatment groups. CONCLUSION The present study demonstrates that rivaroxaban can attenuate AD progress and can be a potential choice to prevent AD.
Collapse
Affiliation(s)
- Zhihong Bian
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Xia Liu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Tian Feng
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Haibo Yu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Xiao Hu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Xinran Hu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Yuting Bian
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Hongming Sun
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Koh Tadokoro
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Mami Takemoto
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Taijun Yunoki
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Yumiko Nakano
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Yusuke Fukui
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Ryuta Morihara
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Koji Abe
- National Center Hospital, National Center of Neurology and Psychiatry, Kodaira-shi, Tokyo, Japan
| | - Toru Yamashita
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| |
Collapse
|
4
|
Triplet EM, Kim HN, Yoon H, Radulovic M, Kleppe L, Simon WL, Choi CI, Walsh PJ, Dutton JR, Scarisbrick IA. The thrombin receptor links brain derived neurotrophic factor to neuron cholesterol production, resiliency and repair after spinal cord injury. Neurobiol Dis 2021; 152:105294. [PMID: 33549720 PMCID: PMC8021459 DOI: 10.1016/j.nbd.2021.105294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/13/2021] [Accepted: 02/03/2021] [Indexed: 11/28/2022] Open
Abstract
Despite concerted efforts to identify CNS regeneration strategies, an incomplete understanding of how the needed molecular machinery is regulated limits progress. Here we use models of lateral compression and FEJOTA clip contusion-compression spinal cord injury (SCI) to identify the thrombin receptor (Protease Activated Receptor 1 (PAR1)) as an integral facet of this machine with roles in regulating neurite growth through a growth factor- and cholesterol-dependent mechanism. Functional recovery and signs of neural repair, including expression of cholesterol biosynthesis machinery and markers of axonal and synaptic integrity, were all increased after SCI in PAR1 knockout female mice, while PTEN was decreased. Notably, PAR1 differentially regulated HMGCS1, a gene encoding a rate-limiting enzyme in cholesterol production, across the neuronal and astroglial compartments of the intact versus injured spinal cord. Pharmacologic inhibition of cortical neuron PAR1 using vorapaxar in vitro also decreased PTEN and promoted neurite outgrowth in a cholesterol dependent manner, including that driven by suboptimal brain derived neurotrophic factor (BDNF). Pharmacologic inhibition of PAR1 also augmented BDNF-driven HMGCS1 and cholesterol production by murine cortical neurons and by human SH-SY5Y and iPSC-derived neurons. The link between PAR1, cholesterol and BDNF was further highlighted by demonstrating that the deleterious effects of PAR1 over-activation are overcome by supplementing cultures with BDNF, cholesterol or by blocking an inhibitor of adenylate cyclase, Gαi. These findings document PAR1-linked neurotrophic coupling mechanisms that regulate neuronal cholesterol metabolism as an important component of the machinery regulating CNS repair and point to new strategies to enhance neural resiliency after injury.
Collapse
Affiliation(s)
- Erin M Triplet
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic Alix School of Medicine and the Mayo Clinic Medical Scientist Training Program Sciences Rochester, United States of America
| | - Ha Neui Kim
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, United States of America
| | - Hyesook Yoon
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, United States of America
| | - Maja Radulovic
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, United States of America
| | - Laurel Kleppe
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, United States of America
| | - Whitney L Simon
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, United States of America
| | - Chan-Il Choi
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, United States of America
| | - Patrick J Walsh
- Department of Genetics and Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - James R Dutton
- Department of Genetics and Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Isobel A Scarisbrick
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic Alix School of Medicine and the Mayo Clinic Medical Scientist Training Program Sciences Rochester, United States of America; Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, United States of America; Department of Physiology and Biomedical Engineering, Rochester, MN 55905, United States of America.
| |
Collapse
|
5
|
Billi AC, Ludwig JE, Fritz Y, Rozic R, Swindell WR, Tsoi LC, Gruzska D, Abdollahi-Roodsaz S, Xing X, Diaconu D, Uppala R, Camhi MI, Klenotic PA, Sarkar MK, Husni ME, Scher JU, McDonald C, Kahlenberg JM, Midura RJ, Gudjonsson JE, Ward NL. KLK6 expression in skin induces PAR1-mediated psoriasiform dermatitis and inflammatory joint disease. J Clin Invest 2021; 130:3151-3157. [PMID: 32155135 DOI: 10.1172/jci133159] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 02/27/2020] [Indexed: 12/12/2022] Open
Abstract
Kallikrein-related peptidase 6 (KLK6) is a secreted serine protease hypothesized to promote inflammation via cleavage of protease-activated receptor 1 (PAR1) and PAR2. KLK6 levels are elevated in multiple inflammatory and autoimmune conditions, but no definitive role in pathogenesis has been established. Here, we show that skin-targeted overexpression of KLK6 causes generalized, severe psoriasiform dermatitis with spontaneous development of debilitating psoriatic arthritis-like joint disease. The psoriatic skin and joint phenotypes are reversed by normalization of skin KLK6 levels and attenuated following genetic elimination of PAR1 but not PAR2. Conservation of this regulatory pathway was confirmed in human psoriasis using vorapaxar, an FDA-approved PAR1 antagonist, on explanted lesional skin from patients with psoriasis. Beyond defining a critical role for KLK6/PAR1 signaling in promoting psoriasis, our results demonstrate that KLK6/PAR1-mediated inflammation in the skin alone is sufficient to drive inflammatory joint disease. Further, we identify PAR1 as a promising cytokine-independent target in therapy of psoriasis and psoriatic arthritis.
Collapse
Affiliation(s)
- Allison C Billi
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jessica E Ludwig
- Department of Dermatology and.,Department of Nutrition, Case Western Reserve University, Cleveland, Ohio, USA
| | | | - Richard Rozic
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - William R Swindell
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA.,Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.,Department of Internal Medicine, Jewish Hospital, Cincinnati, Ohio, USA
| | - Lam C Tsoi
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA.,Center for Statistical Genetics, Department of Biostatistics, and.,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Dennis Gruzska
- Department of Dermatology and.,Department of Nutrition, Case Western Reserve University, Cleveland, Ohio, USA
| | - Shahla Abdollahi-Roodsaz
- Inflammation and Immunology Thematic Center of Excellence, Celgene Corp., Cambridge, Massachusetts, USA
| | - Xianying Xing
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Ranjitha Uppala
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | - Mrinal K Sarkar
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - M Elaine Husni
- Department of Rheumatologic and Immunologic Disease, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jose U Scher
- Department of Medicine, New York University, New York, New York, USA
| | - Christine McDonald
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Ronald J Midura
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Nicole L Ward
- Department of Dermatology and.,Department of Nutrition, Case Western Reserve University, Cleveland, Ohio, USA.,Murdough Family Center for Psoriasis, Cleveland, Ohio, USA
| |
Collapse
|
6
|
Spinal PAR2 Activation Contributes to Hypersensitivity Induced by Peripheral Inflammation in Rats. Int J Mol Sci 2021; 22:ijms22030991. [PMID: 33498178 PMCID: PMC7863954 DOI: 10.3390/ijms22030991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/16/2021] [Accepted: 01/17/2021] [Indexed: 11/17/2022] Open
Abstract
The mechanisms of inflammatory pain need to be identified in order to find new superior treatments. Protease-activated receptors 2 (PAR2) and transient receptor potential vanilloid 1 (TRPV1) are highly co-expressed in dorsal root ganglion neurons and implicated in pain development. Here, we examined the role of spinal PAR2 in hyperalgesia and the modulation of synaptic transmission in carrageenan-induced peripheral inflammation, using intrathecal (i.t.) treatment in the behavioral experiments and recordings of spontaneous, miniature and dorsal root stimulation-evoked excitatory postsynaptic currents (sEPSCs, mEPSCs and eEPSCs) in spinal cord slices. Intrathecal PAR2-activating peptide (AP) administration aggravated the carrageenan-induced thermal hyperalgesia, and this was prevented by a TRPV1 antagonist (SB 366791) and staurosporine i.t. pretreatment. Additionally, the frequency of the mEPSC and sEPSC and the amplitude of the eEPSC recorded from the superficial dorsal horn neurons were enhanced after acute PAR2 AP application, while prevented with SB 366791 or staurosporine pretreatment. PAR2 antagonist application reduced the thermal hyperalgesia and decreased the frequency of mEPSC and sEPSC and the amplitude of eEPSC. Our findings highlight the contribution of spinal PAR2 activation to carrageenan-induced hyperalgesia and the importance of dorsal horn PAR2 and TRPV1 receptor interactions in the modulation of nociceptive synaptic transmission.
Collapse
|
7
|
Brenner M, Messing A. Regulation of GFAP Expression. ASN Neuro 2021; 13:1759091420981206. [PMID: 33601918 PMCID: PMC7897836 DOI: 10.1177/1759091420981206] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022] Open
Abstract
Expression of the GFAP gene has attracted considerable attention because its onset is a marker for astrocyte development, its upregulation is a marker for reactive gliosis, and its predominance in astrocytes provides a tool for their genetic manipulation. The literature on GFAP regulation is voluminous, as almost any perturbation of development or homeostasis in the CNS will lead to changes in its expression. In this review, we limit our discussion to mechanisms proposed to regulate GFAP synthesis through a direct interaction with its gene or mRNA. Strengths and weaknesses of the supportive experimental findings are described, and suggestions made for additional studies. This review covers 15 transcription factors, DNA and histone methylation, and microRNAs. The complexity involved in regulating the expression of this intermediate filament protein suggests that GFAP function may vary among both astrocyte subtypes and other GFAP-expressing cells, as well as during development and in response to perturbations.
Collapse
Affiliation(s)
- Michael Brenner
- Department of Neurobiology, University of Alabama-Birmingham, Birmingham, Alabama, United States
| | - Albee Messing
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, United States
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
| |
Collapse
|
8
|
Li Z, Song Y, He T, Wen R, Li Y, Chen T, Huang S, Wang Y, Tang Y, Shen F, Tian HL, Yang GY, Zhang Z. M2 microglial small extracellular vesicles reduce glial scar formation via the miR-124/STAT3 pathway after ischemic stroke in mice. Am J Cancer Res 2021; 11:1232-1248. [PMID: 33391532 PMCID: PMC7738903 DOI: 10.7150/thno.48761] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/01/2020] [Indexed: 12/23/2022] Open
Abstract
Rationale: Glial scars present a major obstacle for neuronal regeneration after stroke. Thus, approaches to promote their degradation and inhibit their formation are beneficial for stroke recovery. The interaction of microglia and astrocytes is known to be involved in glial scar formation after stroke; however, how microglia affect glial scar formation remains unclear. Methods: Mice were treated daily with M2 microglial small extracellular vesicles through tail intravenous injections from day 1 to day 7 after middle cerebral artery occlusion. Glial scar, infarct volume, neurological score were detected after ischemia. microRNA and related protein were examined in peri-infarct areas of the brain following ischemia. Results: M2 microglial small extracellular vesicles reduced glial scar formation and promoted recovery after stroke and were enriched in miR-124. Furthermore, M2 microglial small extracellular vesicle treatment decreased the expression of the astrocyte proliferation gene signal transducer and activator of transcription 3, one of the targets of miR-124, and glial fibrillary acidic protein and inhibited astrocyte proliferation both in vitro and in vivo. It also decreased Notch 1 expression and increased Sox2 expression in astrocytes, which suggested that astrocytes had transformed into neuronal progenitor cells. Finally, miR-124 knockdown in M2 microglial small extracellular vesicles blocked their effects on glial scars and stroke recovery. Conclusions: Our results showed, for the first time, that microglia regulate glial scar formation via small extracellular vesicles, indicating that M2 microglial small extracellular vesicles could represent a new therapeutic approach for stroke.
Collapse
|
9
|
Protein Degradome of Spinal Cord Injury: Biomarkers and Potential Therapeutic Targets. Mol Neurobiol 2020; 57:2702-2726. [PMID: 32328876 DOI: 10.1007/s12035-020-01916-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 03/31/2020] [Indexed: 12/13/2022]
Abstract
Degradomics is a proteomics sub-discipline whose goal is to identify and characterize protease-substrate repertoires. With the aim of deciphering and characterizing key signature breakdown products, degradomics emerged to define encryptic biomarker neoproteins specific to certain disease processes. Remarkable improvements in structural and analytical experimental methodologies as evident in research investigating cellular behavior in neuroscience and cancer have allowed the identification of specific degradomes, increasing our knowledge about proteases and their regulators and substrates along with their implications in health and disease. A physiologic balance between protein synthesis and degradation is sought with the activation of proteolytic enzymes such as calpains, caspases, cathepsins, and matrix metalloproteinases. Proteolysis is essential for development, growth, and regeneration; however, inappropriate and uncontrolled activation of the proteolytic system renders the diseased tissue susceptible to further neurotoxic processes. In this article, we aim to review the protease-substrate repertoires as well as emerging therapeutic interventions in spinal cord injury at the degradomic level. Several protease substrates and their breakdown products, essential for the neuronal structural integrity and functional capacity, have been characterized in neurotrauma including cytoskeletal proteins, neuronal extracellular matrix glycoproteins, cell junction proteins, and ion channels. Therefore, targeting exaggerated protease activity provides a potentially effective therapeutic approach in the management of protease-mediated neurotoxicity in reducing the extent of damage secondary to spinal cord injury.
Collapse
|
10
|
Rajput PS, Lamb J, Kothari S, Pereira B, Soetkamp D, Wang Y, Tang J, Van Eyk JE, Mullins ES, Lyden PD. Neuron-generated thrombin induces a protective astrocyte response via protease activated receptors. Glia 2019; 68:246-262. [PMID: 31453648 DOI: 10.1002/glia.23714] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 08/02/2019] [Accepted: 08/15/2019] [Indexed: 01/08/2023]
Abstract
Astrocytes protect neurons during cerebral injury through several postulated mechanisms. Recent therapeutic attention has focused on enhancing or augmenting the neuroprotective actions of astrocytes but in some instances astrocytes can assume a neurotoxic phenotype. The signaling mechanisms that drive astrocytes toward a protective versus toxic phenotype are not fully known but cell-cell signaling via proteases acting on cell-specific receptors underlies critical mechanistic steps in neurodevelopment and disease. The protease activated receptor (PAR), resides in multiple brain cell types, and most PARs are found on astrocytes. We asked whether neuron-generated thrombin constituted an important astrocyte activation signal because our previous studies have shown that neurons contain prothrombin gene and transcribed protein. We used neuron and astrocyte mono-cell cultures exposed to oxygen-glucose deprivation and a model of middle cerebral artery occlusion. We found that ischemic neurons secrete thrombin into culture media, which leads to astrocyte activation; such astrocyte activation can be reproduced with low doses of thrombin. Media from prothrombin-deficient neurons failed to activate astrocytes and adding thrombin to such media restored activation. Astrocytes lacking PAR1 did not respond to neuron-generated thrombin. Induced astrocyte activation was antagonized dose-dependently with thrombin inhibitors or PAR1 antagonists. Ischemia-induced astrocyte activation in vivo was inhibited after neuronal prothrombin knockout, resulting in larger strokes. Restoring prothrombin to neurons with a lentiviral gene vector restored astrocyte activation and reduced stroke damage. We conclude that neuron-generated thrombin, released during ischemia, acts via PAR1 and may cause astrocyte activation and paracrine neuroprotection.
Collapse
Affiliation(s)
- Padmesh S Rajput
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jessica Lamb
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Shweta Kothari
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Benedict Pereira
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Daniel Soetkamp
- The Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Yizhou Wang
- Genomics Core, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jie Tang
- Genomics Core, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jennifer E Van Eyk
- The Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Eric S Mullins
- Division of Hematology and Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Patrick D Lyden
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
11
|
Yoon H, Radulovic M, Scarisbrick IA. Kallikrein-related peptidase 6 orchestrates astrocyte form and function through proteinase activated receptor-dependent mechanisms. Biol Chem 2019; 399:1041-1052. [PMID: 29604205 DOI: 10.1515/hsz-2018-0122] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/26/2018] [Indexed: 02/01/2023]
Abstract
Kallikrein-related peptidase 6 (Klk6) is the most abundant serine proteinase in the adult central nervous system (CNS), yet we know little regarding its physiological roles or mechanisms of action. Levels of Klk6 in the extracellular environment are dynamically regulated in CNS injury and disease positioning this secreted enzyme to affect cell behavior by potential receptor dependent and independent mechanisms. Here we show that recombinant Klk6 evokes increases in intracellular Ca2+ in primary astrocyte monolayer cultures through activation of proteinase activated receptor 1 (PAR1). In addition, Klk6 promoted a condensation of astrocyte cortical actin leading to an elongated stellate shape and multicellular aggregation in a manner that was dependent on the presence of either PAR1 or PAR2. Klk6-evoked changes in astrocyte shape were accompanied by translocation of β-catenin from the plasma membrane to the cytoplasm. These data are exciting because they demonstrate that Klk6 can influence astrocyte plasticity through receptor-dependent mechanisms. Furthermore, this study expands our understanding of the mechanisms by which kallikreins can contribute to neural homeostasis and remodeling and point to both PAR1 and PAR2 as new therapeutic targets to modulate astrocyte form and function.
Collapse
Affiliation(s)
- Hyesook Yoon
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN 55905, USA.,Rehabilitation Medicine Research Center, Mayo Clinic, 200 First St., SW, Rochester, MN 55905, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Maja Radulovic
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN 55905, USA.,Rehabilitation Medicine Research Center, Mayo Clinic, 200 First St., SW, Rochester, MN 55905, USA
| | - Isobel A Scarisbrick
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN 55905, USA.,Rehabilitation Medicine Research Center, Mayo Clinic, 200 First St., SW, Rochester, MN 55905, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
12
|
Choi CI, Yoon H, Drucker KL, Langley MR, Kleppe L, Scarisbrick IA. The Thrombin Receptor Restricts Subventricular Zone Neural Stem Cell Expansion and Differentiation. Sci Rep 2018; 8:9360. [PMID: 29921916 PMCID: PMC6008392 DOI: 10.1038/s41598-018-27613-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/06/2018] [Indexed: 01/05/2023] Open
Abstract
Thrombin is frequently increased in the CNS after injury yet little is known regarding its effects on neural stem cells. Here we show that the subventricular zone (SVZ) of adult mice lacking the high affinity receptor for thrombin, proteinase activated receptor 1 (PAR1), show increased numbers of Sox2+ and Ki-67+ self-renewing neural stem cells (NSCs) and Olig2+ oligodendrocyte progenitors. SVZ NSCs derived from PAR1-knockout mice, or treated with a PAR1 small molecule inhibitor (SCH79797), exhibited enhanced capacity for self-renewal in vitro, including increases in neurosphere formation and BrdU incorporation. PAR1-knockout SVZ monolayer cultures contained more Nestin, NG2+ and Olig2+ cells indicative of enhancements in expansion and differentiation towards the oligodendrocyte lineage. Cultures of NSCs lacking PAR1 also expressed higher levels of myelin basic protein, proteolipid protein and glial fibrillary acidic protein upon differentiation. Complementing these findings, the corpus callosum and anterior commissure of adult PAR1-knockout mice contained greater numbers of Olig2+ progenitors and CC1+ mature oligodendrocytes. Together these findings highlight PAR1 inhibition as a means to expand adult SVZ NSCs and to promote an increased number of mature myelinating oligodendrocytes in vivo that may be of particular benefit in the context of neural injury where PAR1 agonists such as thrombin are deregulated.
Collapse
Affiliation(s)
- Chan-Il Choi
- Department of Physical Medicine and Rehabilitation Mayo Clinic, Rochester, MN, 55905, USA.,Rehabilitation Medicine Research Center Mayo Clinic, Rochester, MN, 55905, USA.,Department of Physiology Mayo Clinic, Rochester, MN, 55905, USA
| | - Hyesook Yoon
- Department of Physical Medicine and Rehabilitation Mayo Clinic, Rochester, MN, 55905, USA.,Rehabilitation Medicine Research Center Mayo Clinic, Rochester, MN, 55905, USA.,Department of Physiology Mayo Clinic, Rochester, MN, 55905, USA
| | - Kristen L Drucker
- Department of Physical Medicine and Rehabilitation Mayo Clinic, Rochester, MN, 55905, USA.,Rehabilitation Medicine Research Center Mayo Clinic, Rochester, MN, 55905, USA
| | - Monica R Langley
- Department of Physical Medicine and Rehabilitation Mayo Clinic, Rochester, MN, 55905, USA.,Rehabilitation Medicine Research Center Mayo Clinic, Rochester, MN, 55905, USA
| | - Laurel Kleppe
- Department of Physical Medicine and Rehabilitation Mayo Clinic, Rochester, MN, 55905, USA.,Rehabilitation Medicine Research Center Mayo Clinic, Rochester, MN, 55905, USA
| | - Isobel A Scarisbrick
- Department of Physical Medicine and Rehabilitation Mayo Clinic, Rochester, MN, 55905, USA. .,Rehabilitation Medicine Research Center Mayo Clinic, Rochester, MN, 55905, USA. .,Department of Physiology Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
13
|
Nokkari A, Abou-El-Hassan H, Mechref Y, Mondello S, Kindy MS, Jaffa AA, Kobeissy F. Implication of the Kallikrein-Kinin system in neurological disorders: Quest for potential biomarkers and mechanisms. Prog Neurobiol 2018; 165-167:26-50. [PMID: 29355711 PMCID: PMC6026079 DOI: 10.1016/j.pneurobio.2018.01.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 01/15/2018] [Indexed: 01/06/2023]
Abstract
Neurological disorders represent major health concerns in terms of comorbidity and mortality worldwide. Despite a tremendous increase in our understanding of the pathophysiological processes involved in disease progression and prevention, the accumulated knowledge so far resulted in relatively moderate translational benefits in terms of therapeutic interventions and enhanced clinical outcomes. Aiming at specific neural molecular pathways, different strategies have been geared to target the development and progression of such disorders. The kallikrein-kinin system (KKS) is among the most delineated candidate systems due to its ubiquitous roles mediating several of the pathophysiological features of these neurological disorders as well as being implicated in regulating various brain functions. Several experimental KKS models revealed that the inhibition or stimulation of the two receptors of the KKS system (B1R and B2R) can exhibit neuroprotective and/or adverse pathological outcomes. This updated review provides background details of the KKS components and their functions in different neurological disorders including temporal lobe epilepsy, traumatic brain injury, stroke, spinal cord injury, Alzheimer's disease, multiple sclerosis and glioma. Finally, this work will highlight the putative roles of the KKS components as potential neurotherapeutic targets and provide future perspectives on the possibility of translating these findings into potential clinical biomarkers in neurological disease.
Collapse
Affiliation(s)
- Amaly Nokkari
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon
| | - Hadi Abou-El-Hassan
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Mark S Kindy
- Department of Pharmaceutical Science, College of Pharmacy, University of South Florida, Tampa, FL, USA; James A. Haley VA Medical Center, Tampa, FL, USA
| | - Ayad A Jaffa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon; Department of Medicine, Medical University of South, Charleston, SC, USA.
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon; Center for Neuroproteomics & Biomarkers Research, Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
14
|
Jones SM, Mann A, Conrad K, Saum K, Hall DE, McKinney LM, Robbins N, Thompson J, Peairs AD, Camerer E, Rayner KJ, Tranter M, Mackman N, Owens AP. PAR2 (Protease-Activated Receptor 2) Deficiency Attenuates Atherosclerosis in Mice. Arterioscler Thromb Vasc Biol 2018; 38:1271-1282. [PMID: 29599135 PMCID: PMC6324171 DOI: 10.1161/atvbaha.117.310082] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 03/15/2018] [Indexed: 12/25/2022]
Abstract
OBJECTIVE PAR2 (protease-activated receptor 2)-dependent signaling results in augmented inflammation and has been implicated in the pathogenesis of several autoimmune conditions. The objective of this study was to determine the effect of PAR2 deficiency on the development of atherosclerosis. APPROACH AND RESULTS PAR2 mRNA and protein expression is increased in human carotid artery and mouse aortic arch atheroma versus control carotid and aortic arch arteries, respectively. To determine the effect of PAR2 deficiency on atherosclerosis, male and female low-density lipoprotein receptor-deficient (Ldlr-/-) mice (8-12 weeks old) that were Par2+/+ or Par2-/- were fed a fat- and cholesterol-enriched diet for 12 or 24 weeks. PAR2 deficiency attenuated atherosclerosis in the aortic sinus and aortic root after 12 and 24 weeks. PAR2 deficiency did not alter total plasma cholesterol concentrations or lipoprotein distributions. Bone marrow transplantation showed that PAR2 on nonhematopoietic cells contributed to atherosclerosis. PAR2 deficiency significantly attenuated levels of the chemokines Ccl2 and Cxcl1 in the circulation and macrophage content in atherosclerotic lesions. Mechanistic studies using isolated primary vascular smooth muscle cells showed that PAR2 deficiency is associated with reduced Ccl2 and Cxcl1 mRNA expression and protein release into the supernatant resulting in less monocyte migration. CONCLUSIONS Our results indicate that PAR2 deficiency is associated with attenuation of atherosclerosis and may reduce lesion progression by blunting Ccl2- and Cxcl1-induced monocyte infiltration.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Aortic Diseases/prevention & control
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Carotid Artery Diseases/genetics
- Carotid Artery Diseases/metabolism
- Carotid Artery Diseases/pathology
- Cell Movement
- Cells, Cultured
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Chemokine CXCL1/genetics
- Chemokine CXCL1/metabolism
- Disease Models, Animal
- Female
- Genetic Predisposition to Disease
- Humans
- Lipids/blood
- Macrophages/metabolism
- Macrophages/pathology
- Male
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Plaque, Atherosclerotic
- Receptor, PAR-1/deficiency
- Receptor, PAR-1/genetics
- Receptor, PAR-2/deficiency
- Receptor, PAR-2/genetics
- Receptors, G-Protein-Coupled/deficiency
- Receptors, G-Protein-Coupled/genetics
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
Collapse
Affiliation(s)
- Shannon M Jones
- From the Division of Cardiovascular Health and Disease (S.M.J., A.M., K.C., K.S., L.M.M., N.R., M.T., A.P.O.)
| | - Adrien Mann
- From the Division of Cardiovascular Health and Disease (S.M.J., A.M., K.C., K.S., L.M.M., N.R., M.T., A.P.O.)
| | - Kelsey Conrad
- From the Division of Cardiovascular Health and Disease (S.M.J., A.M., K.C., K.S., L.M.M., N.R., M.T., A.P.O.)
- Pathobiology and Molecular Medicine Program (K.C., M.T., A.P.O.)
| | - Keith Saum
- From the Division of Cardiovascular Health and Disease (S.M.J., A.M., K.C., K.S., L.M.M., N.R., M.T., A.P.O.)
- University of Cincinnati Medical Scientist Training Program (K.S.)
| | - David E Hall
- Department of Nutritional Sciences, College of Allied Health (D.E.H., A.D.P.)
- Department of Internal Medicine (D.E.H., A.D.P.), University of Cincinnati College of Medicine, OH
| | - Lisa M McKinney
- From the Division of Cardiovascular Health and Disease (S.M.J., A.M., K.C., K.S., L.M.M., N.R., M.T., A.P.O.)
| | - Nathan Robbins
- From the Division of Cardiovascular Health and Disease (S.M.J., A.M., K.C., K.S., L.M.M., N.R., M.T., A.P.O.)
| | - Joel Thompson
- Division of Endocrinology and Molecular Medicine, Department of Internal Medicine, University of Kentucky, Lexington (J.T.)
| | - Abigail D Peairs
- Department of Nutritional Sciences, College of Allied Health (D.E.H., A.D.P.)
- Department of Internal Medicine (D.E.H., A.D.P.), University of Cincinnati College of Medicine, OH
| | - Eric Camerer
- INSERM U970, Paris Cardiovascular Research Centre, France (E.C.)
| | - Katey J Rayner
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Michael Tranter
- From the Division of Cardiovascular Health and Disease (S.M.J., A.M., K.C., K.S., L.M.M., N.R., M.T., A.P.O.)
- Pathobiology and Molecular Medicine Program (K.C., M.T., A.P.O.)
| | - Nigel Mackman
- Division of Hematology and Oncology, Department of Medicine, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill (N.M.)
| | - A Phillip Owens
- From the Division of Cardiovascular Health and Disease (S.M.J., A.M., K.C., K.S., L.M.M., N.R., M.T., A.P.O.)
- Pathobiology and Molecular Medicine Program (K.C., M.T., A.P.O.)
| |
Collapse
|
15
|
Agrawal K, Arora N. Serine protease allergen favours Th2 responses via PAR-2 and STAT3 activation in murine model. Allergy 2018; 73:569-575. [PMID: 28940472 DOI: 10.1111/all.13315] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Protease activity of Per a 10 favours Th2 responses by differential regulation of IL-12p70 and IL-23 cytokine subunits. This study aimed to elucidate the underlying mechanism of differential regulation of IL-12p70 and IL-23. METHODS PAR-2 activation was blocked in murine model by administering SAM11 before each sensitization. CD11c+ p-STAT3+ cells were measured in lungs by flow cytometry. BMDCs were pretreated with SAM11 or isotype control or stattic and stimulated with Per a 10. p-STAT3 levels were measured using Western blot. Transcript levels of IL-12p35, IL-12/23p40 and IL-23p19 were measured using RT-PCR. Cytokine levels were analysed using ELISA. RESULTS Protease activity of Per a 10 increased p-STAT3 levels in mouse lungs, which was reduced upon PAR-2 blockage. Percentage of p-STAT3+ CD11c+ cells was higher in Per a 10-administered mice and was reduced upon PAR-2 blockage. IL-12p35 and IL-12p70 levels were higher, and IL-23p19 and IL-23 levels were lower in both SAM11-treated mice and BMDCs indicating a role of PAR-2-mediated signalling. IL-4, TSLP, IL-17A, EPO activity, total cell count and specific IgE and IgG1 levels were lower in SAM11-administered mice. Inhibiting STAT3 activation via stattic also leads to lower levels of IL-23p19 and IL-23 and higher levels of IL-12p35. CONCLUSIONS Per a 10 leads to PAR-2 activation on BMDCs resulting in downstream activation of STAT3 to regulate the balance between IL-12/IL-23 subunits causing a cytokine milieu rich in IL-23 to favour Th2 polarization.
Collapse
Affiliation(s)
- K. Agrawal
- Allergy and Immunology section; CSIR-Institute of Genomics and Integrative Biology; New Delhi India
- Academy of Scientific and Innovative Research (AcSIR); CSIR-IGIB Campus; New Delhi India
| | - N. Arora
- Allergy and Immunology section; CSIR-Institute of Genomics and Integrative Biology; New Delhi India
- Academy of Scientific and Innovative Research (AcSIR); CSIR-IGIB Campus; New Delhi India
| |
Collapse
|
16
|
Yoon H, Radulovic M, Walters G, Paulsen AR, Drucker K, Starski P, Wu J, Fairlie DP, Scarisbrick IA. Protease activated receptor 2 controls myelin development, resiliency and repair. Glia 2017; 65:2070-2086. [PMID: 28921694 DOI: 10.1002/glia.23215] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 08/14/2017] [Accepted: 08/16/2017] [Indexed: 12/22/2022]
Abstract
Oligodendrocytes are essential regulators of axonal energy homeostasis and electrical conduction and emerging target cells for restoration of neurological function. Here we investigate the role of protease activated receptor 2 (PAR2), a unique protease activated G protein-coupled receptor, in myelin development and repair using the spinal cord as a model. Results demonstrate that genetic deletion of PAR2 accelerates myelin production, including higher proteolipid protein (PLP) levels in the spinal cord at birth and higher levels of myelin basic protein and thickened myelin sheaths in adulthood. Enhancements in spinal cord myelin with PAR2 loss-of-function were accompanied by increased numbers of Olig2- and CC1-positive oligodendrocytes, as well as in levels of cyclic adenosine monophosphate (cAMP), and extracellular signal related kinase 1/2 (ERK1/2) signaling. Parallel promyelinating effects were observed after blocking PAR2 expression in purified oligodendrocyte cultures, whereas inhibiting adenylate cyclase reversed these effects. Conversely, PAR2 activation reduced PLP expression and this effect was prevented by brain derived neurotrophic factor (BDNF), a promyelinating growth factor that signals through cAMP. PAR2 knockout mice also showed improved myelin resiliency after traumatic spinal cord injury and an accelerated pattern of myelin regeneration after focal demyelination. These findings suggest that PAR2 is an important controller of myelin production and regeneration, both in the developing and adult spinal cord.
Collapse
Affiliation(s)
- Hyesook Yoon
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Rochester, Minnesota, 55905.,Department of Physiology and Biomedical Engineering, Rochester, Minnesota, 55905
| | - Maja Radulovic
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Rochester, Minnesota, 55905.,Neurobiology of Disease Program, Mayo Clinic, Rochester, Minnesota, 55905
| | - Grant Walters
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Rochester, Minnesota, 55905
| | - Alex R Paulsen
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Rochester, Minnesota, 55905
| | - Kristen Drucker
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Rochester, Minnesota, 55905
| | - Phillip Starski
- Neurobiology of Disease Program, Mayo Clinic, Rochester, Minnesota, 55905
| | - Jianmin Wu
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Rochester, Minnesota, 55905
| | - David P Fairlie
- ARC Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Isobel A Scarisbrick
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Rochester, Minnesota, 55905.,Department of Physiology and Biomedical Engineering, Rochester, Minnesota, 55905.,Neurobiology of Disease Program, Mayo Clinic, Rochester, Minnesota, 55905
| |
Collapse
|
17
|
Yoon H, Walters G, Paulsen AR, Scarisbrick IA. Astrocyte heterogeneity across the brain and spinal cord occurs developmentally, in adulthood and in response to demyelination. PLoS One 2017; 12:e0180697. [PMID: 28700615 PMCID: PMC5507262 DOI: 10.1371/journal.pone.0180697] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/20/2017] [Indexed: 01/08/2023] Open
Abstract
Astrocytes have emerged as essential regulators of function and response to injury in the brain and spinal cord, yet very little is known about regional differences that exist. Here we compare the expression of key astroglial markers (glial fibrillary acidic protein (GFAP) and Aldehyde Dehydrogenase-1 Family Member L1 (ALDH1L1)) across these disparate poles of the neuraxis, tracking their expression developmentally and in the context of demyelination. In addition, we document changes in the astrocyte regulatory cytokine interleukin 6 (IL-6), and its signaling partner signal transducer and activator of transcription 3 (STAT3), in vivo and in vitro. Results demonstrate that GFAP expression is higher in the developing and adult spinal cord relative to brain. Comparisons between GFAP and ALDH1L1 expression suggest elevations in spinal cord GFAP during the early postnatal period reflect an accelerated appearance of astrocytes, while elevations in adulthood reflect higher expression by individual astrocytes. Notably, increases in spinal cord compared to whole brain GFAP were paralleled by higher levels of IL-6 and STAT3. Equivalent elevations in GFAP, GFAP/ALDH1L1 ratios, and in IL-6, were observed in primary astrocyte cultures derived from spinal cord compared to cortex. Also, higher levels of GFAP were observed in the spinal cord compared to the brain after focal demyelinating injury. Altogether, these studies point to key differences in astrocyte abundance and the expression of GFAP and IL-6 across the brain and spinal cord that are positioned to influence regional specialization developmentally and responses occurring in the context of injury and disease.
Collapse
Affiliation(s)
- Hyesook Yoon
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Grant Walters
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Alex R. Paulsen
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Isobel A. Scarisbrick
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States of America
- Neurobiology of Disease Program, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
18
|
Yoon H, Scarisbrick IA. Kallikrein-related peptidase 6 exacerbates disease in an autoimmune model of multiple sclerosis. Biol Chem 2017; 397:1277-1286. [PMID: 27533119 DOI: 10.1515/hsz-2016-0239] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/10/2016] [Indexed: 12/22/2022]
Abstract
Kallikrein-related peptidase 6 (Klk6) is elevated in the serum of multiple sclerosis (MS) patients and is hypothesized to participate in inflammatory and neuropathogenic aspects of the disease. To test this hypothesis, we investigated the impact of systemic administration of recombinant Klk6 on the development and progression of MOG35-55-induced experimental autoimmune encephalomyelitis (EAE). First, we determined that Klk6 expression is elevated in the spinal cord of mice with EAE at the peak of clinical disease and in immune cells upon priming with the disease-initiating peptide in vitro. Systemic administration of recombinant Klk6 to mice during the priming phase of disease resulted in an exacerbation of clinical symptoms, including earlier onset of disease and higher levels of spinal cord inflammation and pathology. Treatment of MOG35-55-primed immune cells with Klk6 in culture enhanced expression of pro-inflammatory cytokines, interferon-γ, tumor necrosis factor, and interleukin-17, while reducing anti-inflammatory cytokines interleukin-4 and interleukin-5. Together these findings provide evidence that elevations in systemic Klk6 can bias the immune system towards pro-inflammatory responses capable of exacerbating the development of neuroinflammation and paralytic neurological deficits. We suggest that Klk6 represents an important target for conditions in which pro-inflammatory responses play a critical role in disease development, including MS.
Collapse
|
19
|
Li LM, Han M, Jiang XC, Yin XZ, Chen F, Zhang TY, Ren H, Zhang JW, Hou TJ, Chen Z, Ou-Yang HW, Tabata Y, Shen YQ, Gao JQ. Peptide-Tethered Hydrogel Scaffold Promotes Recovery from Spinal Cord Transection via Synergism with Mesenchymal Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2017; 9:3330-3342. [PMID: 28058831 DOI: 10.1021/acsami.6b12829] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Spinal cord injury (SCI) is one of the most devastating injuries. Treatment strategies for SCI are required to overcome comprehensive issues. Implantation of biomaterial scaffolds and stem cells has been demonstrated to be a promising strategy. However, a comprehensive recovery effect is difficult to achieve. In the comprehensive treatment process, the specific roles of the implanted scaffolds and of stem cells in combined strategy are usually neglected. In this study, a peptide-modified scaffold is developed based on hyaluronic acid and an adhesive peptide PPFLMLLKGSTR. Synchrotron radiation micro computed tomography measurement provides insights to the three-dimensional inner topographical property and perspective porous structure of the scaffold. The modified scaffold significantly improves cellular survival and adhesive growth of mesenchymal stem cells during 3D culture in vitro. After implantation in transected spinal cord, the modified scaffold and mesenchymal stems are found to function in synergy to restore injured spinal cord tissue, with respective strengths. Hindlimb motor function scores exhibit the most significant impact of the composite implant at 2 weeks post injury, which is the time secondary injury factors begin to take hold. Investigation on the secondary injury factors including inflammatory response and astrocyte overactivity at 10 days post injury reveals the possible underlying reason. Implants of the scaffold, cells, and especially the combination of both elicit inhibitory effects on these adverse factors. The study develops a promising implant for spinal cord tissue engineering and reveals the roles of the scaffold and stem cells. More importantly, the results provide the first understanding of the bioactive peptide PPFLMLLKGSTR concerning its functions on mesenchymal stem cells and spinal cord tissue restoration.
Collapse
Affiliation(s)
| | | | | | - Xian-Zhen Yin
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201210, China
| | | | | | | | - Ji-Wen Zhang
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201210, China
| | | | | | | | - Yasuhiko Tabata
- Department of Biomaterials, Field of Tissue Engineering, Institute for Frontier Medical Sciences, Kyoto University , Kyoto 606-8507, Japan
| | - You-Qing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University , Hangzhou 310027, China
| | | |
Collapse
|
20
|
Radulovic M, Yoon H, Wu J, Mustafa K, Scarisbrick IA. Targeting the thrombin receptor modulates inflammation and astrogliosis to improve recovery after spinal cord injury. Neurobiol Dis 2016; 93:226-42. [PMID: 27145117 PMCID: PMC4930708 DOI: 10.1016/j.nbd.2016.04.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 04/08/2016] [Accepted: 04/29/2016] [Indexed: 02/07/2023] Open
Abstract
The deregulation of serine protease activity is a common feature of neurological injury, but little is known regarding their mechanisms of action or whether they can be targeted to facilitate repair. In this study we demonstrate that the thrombin receptor (Protease Activated Receptor 1, (PAR1)) serves as a critical translator of the spinal cord injury (SCI) proteolytic microenvironment into a cascade of pro-inflammatory events that contribute to astrogliosis and functional decline. PAR1 knockout mice displayed improved locomotor recovery after SCI and reduced signatures of inflammation and astrogliosis, including expression of glial fibrillary acidic protein (GFAP), vimentin, and STAT3 signaling. SCI-associated elevations in pro-inflammatory cytokines such as IL-1β and IL-6 were also reduced in PAR1-/- mice and co-ordinate improvements in tissue sparing and preservation of NeuN-positive ventral horn neurons, and PKCγ corticospinal axons, were observed. PAR1 and its agonist's thrombin and neurosin were expressed by perilesional astrocytes and each agonist increased the production of IL-6 and STAT3 signaling in primary astrocyte cultures in a PAR1-dependent manner. In turn, IL-6-stimulated astrocytes increased expression of PAR1, thrombin, and neurosin, pointing to a model in which PAR1 activation contributes to increased astrogliosis by feedforward- and feedback-signaling dynamics. Collectively, these findings identify the thrombin receptor as a key mediator of inflammation and astrogliosis in the aftermath of SCI that can be targeted to reduce neurodegeneration and improve neurobehavioral recovery.
Collapse
Affiliation(s)
- Maja Radulovic
- Neurobiology of Disease Program, Mayo Medical and Graduate School, Rehabilitation Medicine Research Center, Rochester 55905, MN, United States
| | - Hyesook Yoon
- Department of Physical Medicine and Rehabilitation, Mayo Medical and Graduate School, Rehabilitation Medicine Research Center, Rochester, MN 55905, United States; Department of Physiology and Biomedical Engineering, Mayo Medical and Graduate School, Rehabilitation Medicine Research Center, Rochester, MN 55905, United States
| | - Jianmin Wu
- Department of Physical Medicine and Rehabilitation, Mayo Medical and Graduate School, Rehabilitation Medicine Research Center, Rochester, MN 55905, United States
| | - Karim Mustafa
- Neurobiology of Disease Program, Mayo Medical and Graduate School, Rehabilitation Medicine Research Center, Rochester 55905, MN, United States
| | - Isobel A Scarisbrick
- Neurobiology of Disease Program, Mayo Medical and Graduate School, Rehabilitation Medicine Research Center, Rochester 55905, MN, United States; Department of Physical Medicine and Rehabilitation, Mayo Medical and Graduate School, Rehabilitation Medicine Research Center, Rochester, MN 55905, United States; Department of Physiology and Biomedical Engineering, Mayo Medical and Graduate School, Rehabilitation Medicine Research Center, Rochester, MN 55905, United States.
| |
Collapse
|
21
|
Cui M, Ma X, Sun J, He J, Shen L, Li F. Effects of STAT3 inhibitors on neural functional recovery after spinal cord injury in rats. Biosci Trends 2016; 10:460-466. [DOI: 10.5582/bst.2016.01160] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Meng Cui
- Department of Orthopaedic Traumatology, Tianjin Hospital
- Tianjin Medical University
| | - Xinlong Ma
- Department of Orthopaedic Traumatology, Tianjin Hospital
| | - Jie Sun
- Department of Orthopaedic Traumatology, Tianjin Hospital
| | - Jinquan He
- Department of Orthopaedic Traumatology, Tianjin Hospital
| | - Lin Shen
- Department of Orthopaedic Traumatology, Tianjin Hospital
| | - Fangguo Li
- Department of Orthopaedic Traumatology, Tianjin Hospital
| |
Collapse
|