1
|
Chen D, Fasina OB, Lin J, Zeng J, Manzoor M, Ohno H, Xiang L, Qi J. TBG096 Ameliorates Memory Deficiency in AD Mouse Model via Promoting Neurogenesis and Regulation of Hsc70/HK2/PKM2/LAMP2A Signaling Pathway. Int J Mol Sci 2025; 26:2804. [PMID: 40141445 PMCID: PMC11943016 DOI: 10.3390/ijms26062804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/07/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
In previous studies, we isolated a series of novel gentisides with nerve growth factor (NGF)-mimic activities from Gentiana rigescens Franch and conducted continuous structure-activity relationship (SAR) studies. Recently, a lead compound named TBG096 was discovered with significant NGF-mimic activity, low toxicity, and ability to pass through the blood-brain barrier (BBB). At the cell level, TBG096 exerts NGF-mimic activity by regulation of heat-shock cognate protein 70 (Hsc70) and downstream proteins. Subsequently, high-fat diet (HFD)-induced Alzheimer disease (AD) mouse models were used to evaluate the anti-AD efficacy of the compound. TBG096 significantly improved the memory dysfunction of AD mice at doses of 0.1, 5, and 20 mg/kg, respectively. In order to elucidate the mechanism of action of the compound against AD, the RNA-sequence analysis of transcriptomics, quantitative real-time polymerase chain reaction (qRT-PCR), immunofluorescence staining, and Western blot analysis were performed using animal samples. TBG096 significantly increased the expression of the Wnt gene family (Wnt10b, Wnt5a, and Wnt1) and the number of mature neurons and newborn neurons in the hippocampus and cerebral cortex of AD mice, respectively. At the same time, it reduced the activity of microglia, astrocyte cells, and expression of inducible nitric oxide synthase (INOS) in the brain. Moreover, this compound significantly increased phosphorylated-adenosine 5'-monophosphate-activated protein kinase (AMPK), Hsc70, and lysosomal-associated membrane protein 2a (LAMP2A) and decreased the expression of hexokinase 2 (HK2), pyruvate kinase M2 (PKM2), amyloid precursor protein (APP), microtubule-associated protein tau (Tau), phosphoryl-Tau, and β-amyloid (Aβ) at the protein level. These results suggest that TBG096 produced the NGF-mimic activity and the anti-AD effect via promoting neurogenesis and modification of the Hsc70/HK2/PKM2/LAMP2A signaling pathway, proposing a potential novel approach to counteracting cognitive decline by developing small molecules that promote neurogenesis and the Hsc70 signaling pathway.
Collapse
Affiliation(s)
- Danni Chen
- College of Pharmaceutical Sciences, Zhejiang University, Yu Hang Tang Road 866, Hangzhou 310058, China; (D.C.); (O.B.F.); (J.L.); (J.Z.); (M.M.)
| | - Opeyemi B. Fasina
- College of Pharmaceutical Sciences, Zhejiang University, Yu Hang Tang Road 866, Hangzhou 310058, China; (D.C.); (O.B.F.); (J.L.); (J.Z.); (M.M.)
| | - Jiahui Lin
- College of Pharmaceutical Sciences, Zhejiang University, Yu Hang Tang Road 866, Hangzhou 310058, China; (D.C.); (O.B.F.); (J.L.); (J.Z.); (M.M.)
| | - Jiayuan Zeng
- College of Pharmaceutical Sciences, Zhejiang University, Yu Hang Tang Road 866, Hangzhou 310058, China; (D.C.); (O.B.F.); (J.L.); (J.Z.); (M.M.)
| | - Majid Manzoor
- College of Pharmaceutical Sciences, Zhejiang University, Yu Hang Tang Road 866, Hangzhou 310058, China; (D.C.); (O.B.F.); (J.L.); (J.Z.); (M.M.)
| | - Hiroshi Ohno
- RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehirocho, Tsutumiku, Yokohama 230-0045, Japan;
| | - Lan Xiang
- College of Pharmaceutical Sciences, Zhejiang University, Yu Hang Tang Road 866, Hangzhou 310058, China; (D.C.); (O.B.F.); (J.L.); (J.Z.); (M.M.)
| | - Jianhua Qi
- College of Pharmaceutical Sciences, Zhejiang University, Yu Hang Tang Road 866, Hangzhou 310058, China; (D.C.); (O.B.F.); (J.L.); (J.Z.); (M.M.)
| |
Collapse
|
2
|
Mitra S, Gera R, Eriksdotter M. NGF-based cholinergic therapies in Alzheimer disease. HANDBOOK OF CLINICAL NEUROLOGY 2025; 211:123-135. [PMID: 40340057 DOI: 10.1016/b978-0-443-19088-9.00007-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
The cholinergic system is part of the parasympathetic nervous system, which works in tandem with the sympathetic and enteric nervous systems to maintain the physiologic functioning of our body. The neurotransmitter acetylcholine (ACh) facilitates cholinergic signaling pathways by activating specific cell surface receptors (nicotinic and muscarinic receptors). Altered cholinergic signaling has been implicated in several pathologic conditions. In this chapter, conditions where cholinergic modulation in the central nervous system occurs through the neurotrophin nerve growth factor (NGF) are addressed. NGF is the master regulator of several pathways, ultimately leading to cell survival, ACh production, regenerative signaling, and anti-inflammatory tone. NGF and cholinergic-related pathways have been reported to be severely affected in the case of Alzheimer disease (AD), the most common dementia disorder. In AD, the cholinergic nuclei of the basal forebrain are affected early during the AD continuum, resulting in cholinergic cell loss and hampered ACh production, which overall affects the propagation of cholinergic signals in other brain regions. Since the 1990s clinically relevant strategies to treat AD patients have been the drugs that enhance cholinergic signaling-termed cholinesterase inhibitors (ChEIs), however, other strategies in AD have been and are presently being assessed for clinical efficacy. Delivery of NGF to the basal forebrain is considered crucial to revive the cholinergic cell bodies, restore ACh production, and sustain cognitive function. This chapter provides a description of the relevance of NGF-based therapies targeted for AD treatment, technical approaches for NGF delivery to the brain, and the status of ongoing clinical studies are provided.
Collapse
Affiliation(s)
- Sumonto Mitra
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Ruchi Gera
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging, Medical Unit Aging, Karolinska University Hospital, Huddinge, Sweden.
| |
Collapse
|
3
|
Napoli D, Orsini N, Salamone G, Calvello MA, Capsoni S, Cattaneo A, Strettoi E. Human NGF "Painless" Ocular Delivery for Retinitis Pigmentosa: An In Vivo Study. eNeuro 2024; 11:ENEURO.0096-24.2024. [PMID: 39293937 PMCID: PMC11412101 DOI: 10.1523/eneuro.0096-24.2024] [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: 02/29/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 09/20/2024] Open
Abstract
Retinitis pigmentosa (RP) is a family of genetically heterogeneous diseases still without a cure. Despite the causative genetic mutation typically not expressed in cone photoreceptors, these cells inevitably degenerate following the primary death of rods, causing blindness. The reasons for the "bystander" degeneration of cones are presently unknown but decrement of survival factors, oxidative stress, and inflammation all play a role. Targeting these generalized biological processes represents a strategy to develop mutation-agnostic therapies for saving vision in large populations of RP individuals. A classical method to support neuronal survival is by employing neurotrophic factors, such as NGF. This study uses painless human NGF (hNGFp), a TrkA receptor-biased variant of the native molecule with lower affinity for nociceptors and limited activity as a pain inducer; the molecule has identical neurotrophic power of the native form but a reduced affinity for the p75NTR receptors, known to trigger apoptosis. hNGFp has a recognized activity on brain microglial cells, which are induced to a phenotype switch from a highly activated to a more homeostatic configuration. hNGFp was administered to RP-like mice in vivo with the aim of decreasing retinal inflammation and also providing retinal neuroprotection. However, the ability of this treatment to counteract the bystander degeneration of cones remained limited.
Collapse
Affiliation(s)
- Debora Napoli
- CNR Neuroscience Institute, Pisa 56124, Italy
- Regional Doctorate School in Neuroscience, University of Florence, Italy
| | - Noemi Orsini
- CNR Neuroscience Institute, Pisa 56124, Italy
- Regional Doctorate School in Neuroscience, University of Florence, Italy
| | | | | | - Simona Capsoni
- Section of Human Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara 44121, Italy
| | - Antonino Cattaneo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
- Rita Levi-Montalcini European Brain Research Institute (EBRI), Roma 00161, Italy
| | | |
Collapse
|
4
|
Capossela L, Gatto A, Ferretti S, Di Sarno L, Graglia B, Massese M, Soligo M, Chiaretti A. Multifaceted Roles of Nerve Growth Factor: A Comprehensive Review with a Special Insight into Pediatric Perspectives. BIOLOGY 2024; 13:546. [PMID: 39056738 PMCID: PMC11273967 DOI: 10.3390/biology13070546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024]
Abstract
Nerve growth factor (NGF) is a neurotrophic peptide largely revealed for its ability to regulate the growth and survival of peripheral sensory, sympathetic, and central cholinergic neurons. The pro-survival and regenerative properties of neurotrophic factors propose a therapeutic potential in a wide range of brain diseases, and NGF, in particular, has appeared as an encouraging potential treatment. In this review, a summary of clinical studies regarding NGF and its therapeutic effects published to date, with a specific interest in the pediatric context, will be attempted. NGF has been studied in neurological disorders such as hypoxic-ischemic encephalopathy, traumatic brain injury, neurobehavioral and neurodevelopmental diseases, congenital malformations, cerebral infections, and in oncological and ocular diseases. The potential of NGF to support neuronal survival, repair, and plasticity in these contexts is highlighted. Emerging therapeutic strategies for NGF delivery, including intranasal administration as well as advanced nanotechnology-based methods, are discussed. These techniques aim to enhance NGF bioavailability and target specificity, optimizing therapeutic outcomes while minimizing systemic side effects. By synthesizing current research, this review underscores the promise and challenges of NGF-based therapies in pediatric neurology, advocating for continued innovation in delivery methods to fully harness NGF's therapeutic potential.
Collapse
Affiliation(s)
- Lavinia Capossela
- Institute of Pediatrics, Fondazione Policlinico A. Gemelli IRCCS-Università Cattolica Sacro Cuore, 00168 Rome, Italy; (S.F.); (L.D.S.); (B.G.); (A.C.)
| | - Antonio Gatto
- Institute of Pediatrics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (A.G.); (M.M.)
| | - Serena Ferretti
- Institute of Pediatrics, Fondazione Policlinico A. Gemelli IRCCS-Università Cattolica Sacro Cuore, 00168 Rome, Italy; (S.F.); (L.D.S.); (B.G.); (A.C.)
| | - Lorenzo Di Sarno
- Institute of Pediatrics, Fondazione Policlinico A. Gemelli IRCCS-Università Cattolica Sacro Cuore, 00168 Rome, Italy; (S.F.); (L.D.S.); (B.G.); (A.C.)
| | - Benedetta Graglia
- Institute of Pediatrics, Fondazione Policlinico A. Gemelli IRCCS-Università Cattolica Sacro Cuore, 00168 Rome, Italy; (S.F.); (L.D.S.); (B.G.); (A.C.)
| | - Miriam Massese
- Institute of Pediatrics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (A.G.); (M.M.)
| | - Marzia Soligo
- Istituto di Farmacologia Traslazionale, Consiglio Nazionale delle Ricerche (CNR), 00133 Rome, Italy;
| | - Antonio Chiaretti
- Institute of Pediatrics, Fondazione Policlinico A. Gemelli IRCCS-Università Cattolica Sacro Cuore, 00168 Rome, Italy; (S.F.); (L.D.S.); (B.G.); (A.C.)
| |
Collapse
|
5
|
Tiberi A, Borgonovo G, Testa G, Pacifico P, Jacob A, Di Caprio M, Totaro V, Calvello M, Cattaneo A, Capsoni S. Reversal of neurological deficits by painless nerve growth factor in a mouse model of Rett syndrome. Brain 2024; 147:122-134. [PMID: 37633263 PMCID: PMC10766238 DOI: 10.1093/brain/awad282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 07/12/2023] [Accepted: 07/31/2023] [Indexed: 08/28/2023] Open
Abstract
Rett syndrome is a rare genetic neurodevelopmental disease, affecting 1 in over 10 000 females born worldwide, caused by de novo mutations in the X-chromosome-located methyl-CpG-binding protein 2 (MeCP2) gene. Despite the great effort put forth by the scientific community, a therapy for this devastating disease is still needed. Here, we tested the therapeutic effects of a painless mutein of the nerve growth factor (NGF), called human NGF painless (hNGFp), via a non-invasive intranasal delivery in female MeCP2+/- mice. Of note, previous work had demonstrated a broad biodistribution of hNGFp in the mouse brain by the nasal delivery route. We report that (i) the long-term lifelong treatment of MeCP2+/- mice with hNGFp, starting at 2 months of age, increased the chance of survival while also greatly improving behavioural parameters. Furthermore, when we assessed the phenotypic changes brought forth by (ii) a short-term 1-month-long hNGFp-treatment, starting at 3 months of age (right after the initial presentation of symptoms), we observed the rescue of a well known neuronal target population of NGF, cholinergic neurons in the medial septum. Moreover, we reveal a deficit in microglial morphology in MeCP2+/- mice, completely reversed in treated animals. This effect on microglia is in line with reports showing microglia to be a TrkA-dependent non-neuronal target cell population of NGF in the brain. To understand the immunomodulatory activity of hNGFp, we analysed the cytokine profile after hNGFp treatment in MeCP2+/- mice, to discover that the treatment recovered the altered expression of key neuroimmune-communication molecules, such as fractalkine. The overall conclusion is that hNGFp delivered intranasally can ameliorate symptoms in the MeCP2+/- model of Rett syndrome, by exerting strong neuroprotection with a dual mechanism of action: directly on target neurons and indirectly via microglia.
Collapse
Affiliation(s)
- Alexia Tiberi
- Institute of Neuroscience, CNR, 56124 Pisa, Italy
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
| | - Giulia Borgonovo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
| | - Giovanna Testa
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
| | - Paola Pacifico
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
| | - Ajesh Jacob
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
| | | | - Valentino Totaro
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
| | | | - Antonino Cattaneo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
- Rita Levi-Montalcini European Brain Research Institute (EBRI), 00161 Roma, Italy
| | - Simona Capsoni
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
- Section of Human Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
6
|
Zhang C, Jiao B, Cao X, Zhang W, Yu S, Zhang K, Zhang M, Zhang X. NTRK1-mediated protection against manganese-induced neurotoxicity and cell apoptosis via IGF2 in SH-SY5Y cells. Biomed Pharmacother 2023; 169:115889. [PMID: 37984302 DOI: 10.1016/j.biopha.2023.115889] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/27/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Excessive manganese (Mn) exposure has been linked to neurotoxicity, cognitive impairments. Neurotrophic Receptor Kinase 1 (NTRK1) encodes Tropomyosin kinase A (TrkA), a neurotrophic receptor, as a mediator of neuron differentiation and survival. Insulin-like growth factor 2 (IGF2), a pivotal member of the insulin gene family, plays a crucial role in brain development and neuroprotection. Despite this knowledge, the precise mechanisms through which NTRK1 and IGF2 influence cell responses to Mn-induced neuronal damage remain elusive. METHODS Cell apoptosis was assessed using CCK8, TUNEL staining, and Western blot analysis of cleaved Caspase-3. Lentiviral vectors facilitated NTRK1 overexpression, while small interfering RNAs (siRNAs) facilitated IGF2 knockdown. Real-time Quantitative PCR (qPCR) determined gene expression levels, while Western blotting measured protein expression. RESULTS The study reveals that NTRK1 inhibits MnCl2-induced apoptosis in SH-SY5Y cells. NTRK1 overexpression significantly upregulated IGF2 expression, and subsequent siRNA-IGF2 experiments confirmed IGF2's pivotal role in NTRK1-mediated neuroprotection. Notably, the study identifies that NTRK1 regulates the expression of IGF2 in the neuroprotective mechanism with the involvement of ER stress pathways. DISCUSSION The study reveals NTRK1's neuroprotective role via IGF2 against Mn-induced neurotoxicity and ER stress modulation in SH-SY5Y cells. These findings offer insights into potential therapies for neurodegenerative disorders related to Mn exposure and NTRK1 dysfunction, driving future research in this domain.
Collapse
Affiliation(s)
- Caixia Zhang
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Bo Jiao
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xueqin Cao
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Wencui Zhang
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Shangchen Yu
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Kaiwen Zhang
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Mi Zhang
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei Province, China.
| | - Xianwei Zhang
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| |
Collapse
|
7
|
Tringali G, Pizzoferrato M, Lisi L, Marinelli S, Buccarello L, Falsini B, Cattaneo A, Navarra P. A Vicious NGF-p75 NTR Positive Feedback Loop Exacerbates the Toxic Effects of Oxidative Damage in the Human Retinal Epithelial Cell Line ARPE-19. Int J Mol Sci 2023; 24:16237. [PMID: 38003427 PMCID: PMC10671591 DOI: 10.3390/ijms242216237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
In spite of its variety of biological activities, the clinical exploitation of human NGF (hNGF) is currently limited to ocular pathologies. It is therefore interesting to test the effects of hNGF in preclinical models that may predict their efficacy and safety in the clinical setting of ocular disorders and compare the effects of hNGF with those of its analogs. We used a human retinal pigment cell line, ARPE-19 cells, to investigate the effects of hNGF and its analogs, mouse NGF (mNGF) and painless NGF (pNGF), on cell viability under basal conditions and after exposure to oxidative stimuli, i.e., hydrogen peroxide (H2O2) and ultraviolet (UV)-A rays. The effects of hNGF and pNGF were also tested on the gene expression and protein synthesis of the two NGF receptor subtypes, p75 neurotrophic receptors (p75NTR) and tyrosine kinase A (TrkA) receptors. We drew the following conclusions: (i) the exposure of ARPE-19 cells to H2O2 or UV-A causes a dose-dependent decrease in the number of viable cells; (ii) under baseline conditions, hNGF, but not pNGF, causes a concentration-dependent decrease in cell viability in the range of doses 1-100 ng/mL; (iii) hNGF, but not pNGF, significantly potentiates the toxic effects of H2O2 or of UV-A on ARPE-19 cells in the range of doses 1-100 ng/mL, while mNGF at the same doses presents an intermediate behavior; (iv) 100 ng/mL of hNGF triggers an increase in p75NTR expression in H2O2-treated ARPE-19 cells, while pNGF at the same dose does not; (v) pNGF, but not hNGF (both given at 100 ng/mL), increases the total cell fluorescence intensity for TrkA receptors in H2O2-treated ARPE-19 cells. The present findings suggest a vicious positive feedback loop through which NGF-mediated upregulation of p75NTR contributes to worsening the toxic effects of oxidative damage in the human retinal epithelial cell line ARPE-19. Looking at the possible clinical relevance of these findings, one can postulate that pNGF might show a better benefit/risk ratio than hNGF in the treatment of ocular disorders.
Collapse
Affiliation(s)
- Giuseppe Tringali
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy (M.P.)
| | - Michela Pizzoferrato
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy (M.P.)
| | - Lucia Lisi
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy (M.P.)
| | - Silvia Marinelli
- European Brain Research Institute-Fondazione Rita Levi Montalcini, 00161 Rome, Italy (L.B.)
| | - Lucia Buccarello
- European Brain Research Institute-Fondazione Rita Levi Montalcini, 00161 Rome, Italy (L.B.)
| | - Benedetto Falsini
- UOC Ophtalmology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- Department of Ophthalmology, Bambino Gesù IRCCS Children’s Hospital, 00133 Rome, Italy
| | - Antonino Cattaneo
- European Brain Research Institute-Fondazione Rita Levi Montalcini, 00161 Rome, Italy (L.B.)
- Bio@SNS Laboratory, Scuola Normale Superiore, 56124 Pisa, Italy
| | - Pierluigi Navarra
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy (M.P.)
| |
Collapse
|
8
|
Tiberi A, Carucci NM, Testa G, Rizzi C, Pacifico P, Borgonovo G, Arisi I, D’Onofrio M, Brandi R, Gan WB, Capsoni S, Cattaneo A. Reduced levels of NGF shift astrocytes toward a neurotoxic phenotype. Front Cell Dev Biol 2023; 11:1165125. [PMID: 37143894 PMCID: PMC10151754 DOI: 10.3389/fcell.2023.1165125] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 03/16/2023] [Indexed: 05/06/2023] Open
Abstract
Nerve growth factor (NGF) is critical for neuronal physiology during development and adulthood. Despite the well-recognized effect of NGF on neurons, less is known about whether NGF can actually affect other cell types in the central nervous system (CNS). In this work, we show that astrocytes are susceptible to changes in ambient levels of NGF. First, we observe that interfering with NGF signaling in vivo via the constitutive expression of an antiNGF antibody induces astrocytic atrophy. A similar asthenic phenotype is encountered in an uncleavable proNGF transgenic mouse model (TgproNGF#72), effectively increasing the brain proNGF levels. To examine whether this effect on astrocytes is cell-autonomous, we cultured wild-type primary astrocytes in the presence of antiNGF antibodies, uncovering that a short incubation period is sufficient to potently and rapidly trigger calcium oscillations. Acute induction of calcium oscillations by antiNGF antibodies is followed by progressive morphological changes similar to those observed in antiNGF AD11 mice. Conversely, incubation with mature NGF has no effect on either calcium activity nor on astrocytic morphology. At longer timescales, transcriptomic analysis revealed that NGF-deprived astrocytes acquire a proinflammatory profile. In particular, antiNGF-treated astrocytes show upregulation of neurotoxic transcripts and downregulation of neuroprotective mRNAs. Consistent with that data, culturing wild-type neurons in the presence of NGF-deprived astrocytes leads to neuronal cell death. Finally, we report that in both awake and anesthetized mice, astrocytes in layer I of the motor cortex respond with an increase in calcium activity to acute NGF inhibition using either NGF-neutralizing antibodies or a TrkA-Fc NGF scavenger. Moreover, in vivo calcium imaging in the cortex of the 5xFAD neurodegeneration mouse model shows an increased level of spontaneous calcium activity in astrocytes, which is significantly reduced after acute administration of NGF. In conclusion, we unveil a novel neurotoxic mechanism driven by astrocytes, triggered by their sensing and reacting to changes in the levels of ambient NGF.
Collapse
Affiliation(s)
- Alexia Tiberi
- BIO@SNS, Scuola Normale Superiore, Pisa, Italy
- Skirball Institute of Biomolecular Medicine, Langone Medical Center, New York University, New York, NY, United States
| | | | | | | | | | | | - Ivan Arisi
- European Brain Research Institute - Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Mara D’Onofrio
- European Brain Research Institute - Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Rossella Brandi
- European Brain Research Institute - Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Wen-Biao Gan
- Skirball Institute of Biomolecular Medicine, Langone Medical Center, New York University, New York, NY, United States
- Shenzhen Bay Laboratory, Shenzhen, China
| | - Simona Capsoni
- BIO@SNS, Scuola Normale Superiore, Pisa, Italy
- Institute of Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Antonino Cattaneo
- BIO@SNS, Scuola Normale Superiore, Pisa, Italy
- European Brain Research Institute - Fondazione Rita Levi-Montalcini, Rome, Italy
- *Correspondence: Antonino Cattaneo,
| |
Collapse
|
9
|
Soligo M, Manni L, Conti G, Chiaretti A. Intranasal nerve growth factor for prevention and recovery of the outcomes of traumatic brain injury. Neural Regen Res 2023; 18:773-778. [DOI: 10.4103/1673-5374.354513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
10
|
Testa G, Mainardi M, Vannini E, Pancrazi L, Cattaneo A, Costa M. Disentangling the signaling complexity of nerve growth factor receptors by
CRISPR
/Cas9. FASEB J 2022; 36:e22498. [PMID: 37036720 DOI: 10.1096/fj.202101760rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 07/25/2022] [Accepted: 08/02/2022] [Indexed: 11/11/2022]
Abstract
The binding of nerve growth factor (NGF) to the tropomyosin-related kinase A (TrkA) and p75NTR receptors activates a large variety of pathways regulating critical processes as diverse as proliferation, differentiation, membrane potential, synaptic plasticity, and pain. To ascertain the details of TrkA-p75NTR interaction and cooperation, a plethora of experiments, mostly based on receptor overexpression or downregulation, have been performed. Among the heterogeneous cellular systems used for studying NGF signaling, the PC12 pheochromocytoma-derived cell line is a widely used model. By means of CRISPR/Cas9 genome editing, we created PC12 cells lacking TrkA, p75NTR , or both. We found that TrkA-null cells become unresponsive to NGF. Conversely, the absence of p75NTR enhances the phosphorylation of TrkA and its effectors. Using a patch-clamp, we demonstrated that the individual activation of TrkA and p75NTR by NGF results in antagonizing effects on the membrane potential. These newly developed PC12 cell lines can be used to investigate the specific roles of TrkA and p75NTR in a genetically defined cellular model, thus providing a useful platform for future studies and further gene editing.
Collapse
Affiliation(s)
- Giovanna Testa
- Laboratory of Biology “Bio@SNS” Scuola Normale Superiore Pisa Italy
| | - Marco Mainardi
- Laboratory of Biology “Bio@SNS” Scuola Normale Superiore Pisa Italy
- Neuroscience Institute National Research Council (CNR) Pisa Italy
| | - Eleonora Vannini
- Neuroscience Institute National Research Council (CNR) Pisa Italy
| | - Laura Pancrazi
- Neuroscience Institute National Research Council (CNR) Pisa Italy
| | - Antonino Cattaneo
- Laboratory of Biology “Bio@SNS” Scuola Normale Superiore Pisa Italy
- European Brain Research Institute “Rita Levi Montalcini” (EBRI) Rome Italy
| | - Mario Costa
- Laboratory of Biology “Bio@SNS” Scuola Normale Superiore Pisa Italy
- Neuroscience Institute National Research Council (CNR) Pisa Italy
- Centro Pisano ricerca e implementazione clinica Flash Radiotherapy “CPFR@CISUP”, “S. Chiara” Hospital Pisa Italy
| |
Collapse
|
11
|
Lisi L, Marinelli S, Ciotti GMP, Pizzoferrato M, Palmerio F, Chiavari M, Cattaneo A, Navarra P. The effects of painless nerve growth factor on human microglia polarization. Front Cell Neurosci 2022; 16:969058. [PMID: 36339818 PMCID: PMC9633670 DOI: 10.3389/fncel.2022.969058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 10/03/2022] [Indexed: 11/26/2023] Open
Abstract
Previous studies in the rat suggest that microglial cells represent a potential druggable target for nerve growth factor (NGF) in the brain. The painless human Nerve Growth Factor (hNGFp) is a recombinant mutated form of human nerve growth factor (hNGF) that shows identical neurotrophic and neuroprotective properties of wild-type NGF but displays at least 10-fold lower algogenic activity. From the pharmacological point of view, hNGFp is a biased tropomyosin receptor kinase A (TrkA) agonist and displays a significantly lower affinity for the p75 neurotrophin receptor (p75NTR). This study aimed to evaluate the expression of TrkA and p75NTR NGF receptors in two different human microglia cell lines, and to investigate the effects of hNGFp and wild-type NGF (NGF) on L-arginine metabolism, taken as a marker of microglia polarization. Both NGF receptors are expressed in human microglia cell lines and are effective in transducing signals triggered by NGF and hNGFp. The latter and, to a lesser extent, NGF inhibit cytokine-stimulated inducible nitric oxide synthase (iNOS) expression and nitric oxide (NO) production in these cells. Conversely NGF but not hNGFp stimulates arginase-mediated urea production.
Collapse
Affiliation(s)
- Lucia Lisi
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Silvia Marinelli
- European Brain Research Institute-Fondazione Rita Levi Montalcini, Rome, Italy
| | - Gabriella Maria Pia Ciotti
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Michela Pizzoferrato
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Federica Palmerio
- European Brain Research Institute-Fondazione Rita Levi Montalcini, Rome, Italy
| | - Marta Chiavari
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Antonino Cattaneo
- European Brain Research Institute-Fondazione Rita Levi Montalcini, Rome, Italy
- Bio@SNS Laboratory, Scuola Normale Superiore, Pisa, Italy
| | - Pierluigi Navarra
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| |
Collapse
|
12
|
Vittorio S, Manelfi C, Gervasoni S, Beccari AR, Pedretti A, Vistoli G, Talarico C. Computational Insights into the Sequence-Activity Relationships of the NGF(1–14) Peptide by Molecular Dynamics Simulations. Cells 2022; 11:cells11182808. [PMID: 36139382 PMCID: PMC9497175 DOI: 10.3390/cells11182808] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/26/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
The Nerve Growth Factor (NGF) belongs to the neurothrophins protein family involved in the survival of neurons in the nervous system. The interaction of NGF with its high-affinity receptor TrkA mediates different cellular pathways related to Alzheimer’s disease, pain, ocular dysfunction, and cancer. Therefore, targeting NGF-TrkA interaction represents a valuable strategy for the development of new therapeutic agents. In recent years, experimental studies have revealed that peptides belonging to the N-terminal domain of NGF are able to partly mimic the biological activity of the whole protein paving the way towards the development of small peptides that can selectively target specific signaling pathways. Hence, understanding the molecular basis of the interaction between the N-terminal segment of NGF and TrkA is fundamental for the rational design of new peptides mimicking the NGF N-terminal domain. In this study, molecular dynamics simulation, binding free energy calculations and per-residue energy decomposition analysis were combined in order to explore the molecular recognition pattern between the experimentally active NGF(1–14) peptide and TrkA. The results highlighted the importance of His4, Arg9 and Glu11 as crucial residues for the stabilization of NGF(1–14)-TrkA interaction, thus suggesting useful insights for the structure-based design of new therapeutic peptides able to modulate NGF-TrkA interaction.
Collapse
Affiliation(s)
- Serena Vittorio
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli, 25, I-20133 Milano, Italy
| | - Candida Manelfi
- Dompé Farmaceutici SpA, EXSCALATE, Via Tommaso De Amicis, 95, I-80131 Napoli, Italy
| | - Silvia Gervasoni
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli, 25, I-20133 Milano, Italy
- Department of Physics, University of Cagliari, I-09042 Monserrato, Italy
| | - Andrea R. Beccari
- Dompé Farmaceutici SpA, EXSCALATE, Via Tommaso De Amicis, 95, I-80131 Napoli, Italy
| | - Alessandro Pedretti
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli, 25, I-20133 Milano, Italy
| | - Giulio Vistoli
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli, 25, I-20133 Milano, Italy
| | - Carmine Talarico
- Dompé Farmaceutici SpA, EXSCALATE, Via Tommaso De Amicis, 95, I-80131 Napoli, Italy
- Correspondence:
| |
Collapse
|
13
|
Li B, Ning B, Yang F, Guo C. Nerve Growth Factor Promotes Retinal Neurovascular Unit Repair: A Review. Curr Eye Res 2022; 47:1095-1105. [PMID: 35499266 DOI: 10.1080/02713683.2022.2055084] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Purpose: The purpose of this paper is to investigate how the imbalance of neurogenic factor (NGF) and its precursor (pro-NGF) mediates structural and functional impairment of retinal neurovascular unit (RNVU) that plays a role in retinal degenerative diseases.Methods: A literature search of electronic databases was performed.Results: The pro-apoptotic effect of pro-NGF and the pro-growth effect of NGF are essential for the pathological and physiological activities of RNVU. Studies show that NGF-based treatment of retinal degenerative diseases, including glaucoma, age-related macular degeneration, retinitis pigmentosa, and diabetic retinopathy, has achieved remarkable efficacy.Conclusions: RNVU plays a complex and multifaceted role in retinal degenerative diseases. The exploration of the differential signaling expression of proNGF-NGF homeostasis under physiological and pathological conditions, and the corresponding pathological processes induced by its regulation, has prompted us to focus on earlier retinal neuroprotective therapeutic strategies to prevent retinal degenerative diseases.
Collapse
Affiliation(s)
- Baohua Li
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, PR China
| | - Bobiao Ning
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, PR China
| | - Fan Yang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, PR China
| | - Chengwei Guo
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, PR China
| |
Collapse
|
14
|
Capsoni S, Cattaneo A. Getting Into the Brain: The Intranasal Approach to Enhance the Delivery of Nerve Growth Factor and Its Painless Derivative in Alzheimer’s Disease and Down Syndrome. Front Neurosci 2022; 16:773347. [PMID: 35360160 PMCID: PMC8961408 DOI: 10.3389/fnins.2022.773347] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 02/10/2022] [Indexed: 01/04/2023] Open
Abstract
The neurotrophin Nerve Growth Factor (NGF) holds a great potential as a therapeutic candidate for the treatment of neurological diseases. However, its safe and effective delivery to the brain is limited by the fact that NGF needs to be selectively targeted to the brain, to avoid severe side effects such as pain and to bypass the blood brain barrier. In this perspective, we will summarize the different approaches that have been used, or are currently applied, to deliver NGF to the brain, during preclinical and clinical trials to develop NGF as a therapeutic drug for Alzheimer’s disease. We will focus on the intranasal delivery of NGF, an approach that is used to deliver proteins to the brain in a non-invasive, safe, and effective manner minimizing systemic exposure. We will also describe the main experimental facts related to the effective intranasal delivery of a mutant form of NGF [painless NGF, human nerve growth factor painless (hNGFp)] in mouse models of Alzheimer’s disease and compare it to other ways to deliver NGF to the brain. We will also report new data on the application of intranasal delivery of hNGFp in Down Syndrome mouse model. These new data extend the therapeutic potential of hNGFp for the treatment of the dementia that is progressively associated to Down Syndrome. In conclusion, we will show how this approach can be a promising strategy and a potential solution for other unmet medical needs of safely and effectively delivering this neuroprotective neurotrophin to the brain.
Collapse
Affiliation(s)
- Simona Capsoni
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
- Section of Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
- *Correspondence: Simona Capsoni,
| | - Antonino Cattaneo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
- European Brain Research Institute–Fondazione Rita Levi-Montalcini, Rome, Italy
| |
Collapse
|
15
|
Manni L, Conti G, Chiaretti A, Soligo M. Intranasal Delivery of Nerve Growth Factor in Neurodegenerative Diseases and Neurotrauma. Front Pharmacol 2021; 12:754502. [PMID: 34867367 PMCID: PMC8635100 DOI: 10.3389/fphar.2021.754502] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/01/2021] [Indexed: 01/01/2023] Open
Abstract
Since the 1980s, the development of a pharmacology based on nerve growth factor (NGF) has been postulated for the therapy of Alzheimer’s disease (AD). This hypothesis was based on the rescuing effect of the neurotrophin on the cholinergic phenotype of the basal forebrain neurons, primarily compromised during the development of AD. Subsequently, the use of NGF was put forward to treat a broader spectrum of neurological conditions affecting the central nervous system, such as Parkinson’s disease, degenerative retinopathies, severe brain traumas and neurodevelopmental dysfunctions. While supported by solid rational assumptions, the progress of a pharmacology founded on these hypotheses has been hampered by the difficulty of conveying NGF towards the brain parenchyma without resorting to invasive and risky delivery methods. At the end of the last century, it was shown that NGF administered intranasally to the olfactory epithelium was able to spread into the brain parenchyma. Notably, after such delivery, pharmacologically relevant concentration of exogenous NGF was found in brain areas located at considerable distances from the injection site along the rostral-caudal axis. These observations paved the way for preclinical characterization and clinical trials on the efficacy of intranasal NGF for the treatment of neurodegenerative diseases and of the consequences of brain trauma. In this review, a summary of the preclinical and clinical studies published to date will be attempted, as well as a discussion about the mechanisms underlying the efficacy and the possible development of the pharmacology based on intranasal conveyance of NGF to the brain.
Collapse
Affiliation(s)
- Luigi Manni
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome, Italy
| | - Giorgio Conti
- Department of Emergency, Intensive Pediatric Therapy and Pediatric Trauma Center, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Chiaretti
- Department of Woman and Child Health, Institute of Pediatrics, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Marzia Soligo
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome, Italy
| |
Collapse
|
16
|
Li F, Geng X, Yun HJ, Haddad Y, Chen Y, Ding Y. Neuroplastic Effect of Exercise Through Astrocytes Activation and Cellular Crosstalk. Aging Dis 2021; 12:1644-1657. [PMID: 34631212 PMCID: PMC8460294 DOI: 10.14336/ad.2021.0325] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/25/2021] [Indexed: 12/21/2022] Open
Abstract
Physical exercise is an effective therapy for neurorehabilitation. Exercise has been shown to induce remodeling and proliferation of astrocyte. Astrocytes potentially affect the recruitment and function of neurons; they could intensify responses of neurons and bring more neurons for the process of neuroplasticity. Interactions between astrocytes, microglia and neurons modulate neuroplasticity and, subsequently, neural circuit function. These cellular interactions promote the number and function of synapses, neurogenesis, and cerebrovascular remodeling. However, the roles and crosstalk of astrocytes with neurons and microglia and any subsequent neuroplastic effects have not been studied extensively in exercise-induced settings. This article discusses the impact of physical exercise on astrocyte proliferation and highlights the interplay between astrocytes, microglia and neurons. The crosstalk between these cells may enhance neuroplasticity, leading to the neuroplastic effects of exercise.
Collapse
Affiliation(s)
- Fengwu Li
- China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China.
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Xiaokun Geng
- China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China.
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, China.
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Ho Jun Yun
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Yazeed Haddad
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Yuhua Chen
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
17
|
Mitra S, Gera R, Linderoth B, Lind G, Wahlberg L, Almqvist P, Behbahani H, Eriksdotter M. A Review of Techniques for Biodelivery of Nerve Growth Factor (NGF) to the Brain in Relation to Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1331:167-191. [PMID: 34453298 DOI: 10.1007/978-3-030-74046-7_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Age-dependent progressive neurodegeneration and associated cognitive dysfunction represent a serious concern worldwide. Currently, dementia accounts for the fifth highest cause of death, among which Alzheimer's disease (AD) represents more than 60% of the cases. AD is associated with progressive cognitive dysfunction which affects daily life of the affected individual and associated family. The cognitive dysfunctions are at least partially due to the degeneration of a specific set of neurons (cholinergic neurons) whose cell bodies are situated in the basal forebrain region (basal forebrain cholinergic neurons, BFCNs) but innervate wide areas of the brain. It has been explicitly shown that the delivery of the neurotrophic protein nerve growth factor (NGF) can rescue BFCNs and restore cognitive dysfunction, making NGF interesting as a potential therapeutic substance for AD. Unfortunately, NGF cannot pass through the blood-brain barrier (BBB) and thus peripheral administration of NGF protein is not viable therapeutically. NGF must be delivered in a way which will allow its brain penetration and availability to the BFCNs to modulate BFCN activity and viability. Over the past few decades, various methodologies have been developed to deliver NGF to the brain tissue. In this chapter, NGF delivery methods are discussed in the context of AD.
Collapse
Affiliation(s)
- Sumonto Mitra
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden.
| | - Ruchi Gera
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Linderoth
- Section of Neurosurgery, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Göran Lind
- Section of Neurosurgery, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Per Almqvist
- Section of Neurosurgery, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Homira Behbahani
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden.,Karolinska Universitets laboratoriet (LNP5), Karolinska University Hospital, Stockholm, Sweden
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
18
|
Testa G, Cattaneo A, Capsoni S. Understanding pain perception through genetic painlessness diseases: The role of NGF and proNGF. Pharmacol Res 2021; 169:105662. [PMID: 34000361 DOI: 10.1016/j.phrs.2021.105662] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/23/2021] [Accepted: 05/03/2021] [Indexed: 01/25/2023]
Abstract
Nerve growth factor (NGF), by binding to TrkA and p75NTR receptors, regulates the survival and differentiation of sensory neurons during development and mediates pain transmission and perception during adulthood, by acting at different levels of the nervous system. Key to understanding the role of NGF as a pain mediator is the finding that mutations (namely, R121W, V232fs and R221W) in the NGF gene cause painlessness disease Hereditary Sensory and Autonomic Neuropathy type V (HSAN V). Here we shall review the consequences of these NGF mutations, each of which results in specific clinical signs: R221W determines congenital pain insensitivity with no overt cognitive disabilities, whereas V232fs and R121W also result in intellectual disability, thus showing similarities to HSAN IV, which is caused by mutations in TrkA, rather than to HSAN V. Comparing the cellular, biochemical and clinical findings of these mutations could help in better understanding not only the possible mechanisms underlying HSAN V, but also mechanisms of NGF signalling and roles. These mutations alter the balance between NGF and proNGF in favour of an accumulation of the latter, suggesting a possible role of proNGF as a molecule with an analgesic role. Furthermore, the neurotrophic and pronociceptive functions of NGF are split by the R221W mutation, making NGF variants based on this mutation interesting for designing therapeutic applications for many diseases. This review emphasizes the possibility of using the mutations involved in "painlessness" clinical disorders as an innovative approach to identify new proteins and pathways involved in pain transmission and perception. OUTSTANDING QUESTIONS: Why do homozygous HSAN V die postnatally? What is the cause of this early postnatal lethality? Is the development of a mouse or a human feeling less pain affecting higher cognitive and perceptual functions? What is the consequence of the HSAN V mutation on the development of joints and bones? Are the multiple fractures observed in HSAN V patients due exclusively to the carelessness consequent to not feeling pain, or also to an intrinsic frailty of their bones? Are heterodimers of NGFWT and NGFR221W in the heterozygote state formed? And if so, what are the properties of these heterodimeric proteins? How is the processing of proNGFR221W to NGFR221W affected by the mutation?
Collapse
Affiliation(s)
- Giovanna Testa
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
| | - Antonino Cattaneo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy.
| | - Simona Capsoni
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy; Section of Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
19
|
La Mendola D, Arena G, Pietropaolo A, Satriano C, Rizzarelli E. Metal ion coordination in peptide fragments of neurotrophins: A crucial step for understanding the role and signaling of these proteins in the brain. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
20
|
Improved neurocognitive performance in FIV infected cats following treatment with the p75 neurotrophin receptor ligand LM11A-31. J Neurovirol 2021; 27:302-324. [PMID: 33661457 DOI: 10.1007/s13365-021-00956-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/31/2020] [Accepted: 02/01/2021] [Indexed: 12/14/2022]
Abstract
HIV rapidly infects the central nervous system (CNS) and establishes a persistent viral reservoir within microglia, perivascular macrophages and astrocytes. Inefficient control of CNS viral replication by antiretroviral therapy results in chronic inflammation and progressive cognitive decline in up to 50% of infected individuals with no effective treatment options. Neurotrophin based therapies have excellent potential to stabilize and repair the nervous system. A novel non-peptide ligand, LM11A-31, that targets the p75 neurotrophin receptor (p75NTR) has been identified as a small bioavailable molecule capable of strong neuroprotection with minimal side effects. To evaluate the neuroprotective effects of LM11A-31 in a natural infection model, we treated cats chronically infected with feline immunodeficiency virus (FIV) with 13 mg/kg LM11A-31 twice daily over a period of 10 weeks and assessed effects on cognitive functions, open field behaviors, activity, sensory thresholds, plasma FIV, cerebrospinal fluid (CSF) FIV, peripheral blood mononuclear cell provirus, CD4 and CD8 cell counts and general physiology. Between 12 and 18 months post-inoculation, cats began to show signs of neural dysfunction in T maze testing and novel object recognition, which were prevented by LM11A-31 treatment. Anxiety-like behavior was reduced in the open field and no changes were seen in sensory thresholds. Systemic FIV titers were unaffected but treated cats exhibited a log drop in CSF FIV titers. No significant adverse effects were observed under all conditions. The data indicate that LM11A-31 is likely to be a potent adjunctive treatment for the control of neurodegeneration in HIV infected individuals.
Collapse
|
21
|
Eriksdotter M, Mitra S. Gene and cell therapy for the nucleus basalis of Meynert with NGF in Alzheimer's disease. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:219-229. [PMID: 34225964 DOI: 10.1016/b978-0-12-819975-6.00012-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
There is currently no effective treatment for the most common of the dementia disorders, Alzheimer's disease (AD). It has been known for decades that the central cholinergic system is important for memory. The cholinergic neurons in the basal forebrain with its cortical and hippocampal projections degenerate in AD and thus contribute to the cognitive decline characteristic of AD. This knowledge led to the development of the currently approved treatment for AD, with inhibitors of acetylcholine-esterase targeting the cholinergic system with beneficial but mild effects. In recent years, other approaches to influence the degenerating cholinergic system in AD focusing on nerve growth factor (NGF) have been undertaken. NGF is required for the survival and function of the basal forebrain cholinergic neurons, the most important being the nucleus basalis of Meynert (nbM). Since there is a lack of NGF and its receptors in the AD forebrain, the hypothesis is that local delivery of NGF to the nbM could revive the cholinergic circuitry and thereby restore cognitive functions. Since NGF does not pass through the blood-brain barrier, approaches involving cerebral injections of genetically modified cells or viral vectors or implantation of encapsulated cells in the nbM in AD patients have been used. These attempts have been partially successful but also have limitations, which are presented and discussed here. In conclusion, these trials point to the importance of further development of NGF-related therapies in AD.
Collapse
Affiliation(s)
- Maria Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden; Theme Aging, Karolinska University Hospital, Huddinge, Sweden.
| | - Sumonto Mitra
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
22
|
Abstract
A major feature of neurodegeneration is disruption of central nervous system homeostasis, during which microglia play diverse roles. In the central nervous system, microglia serve as the first line of immune defense and function in synapse pruning, injury repair, homeostasis maintenance, and regulation of brain development through scavenging and phagocytosis. Under pathological conditions or various stimulations, microglia proliferate, aggregate, and undergo a variety of changes in cell morphology, immunophenotype, and function. This review presents the features of microglia, especially their diversity and ability to change dynamically, and reinterprets their role as sensors for multiple stimulations and as effectors for brain aging and neurodegeneration. This review also summarizes some therapeutic approaches for neurodegenerative diseases that target microglia.
Collapse
Affiliation(s)
- Yu Xu
- Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Embryo Original Disease, Shanghai Municipal Key Clinical Specialty; Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming-Zhu Jin
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ze-Yong Yang
- Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Embryo Original Disease, Shanghai Municipal Key Clinical Specialty, Shanghai, China
| | - Wei-Lin Jin
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering; National Centers for Translational Medicine, Shanghai Jiao Tong University, Shanghai; Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
23
|
Yang W, Sung K, Xu W, Rodriguez MJ, Wu AC, Santos SA, Fang S, Uber RK, Dong SX, Guillory BC, Orain X, Raus J, Jolivalt C, Calcutt N, Rissman RA, Ding J, Wu C. A missense point mutation in nerve growth factor (NGF R100W) results in selective peripheral sensory neuropathy. Prog Neurobiol 2020; 194:101886. [PMID: 32693191 DOI: 10.1016/j.pneurobio.2020.101886] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 06/09/2020] [Accepted: 07/11/2020] [Indexed: 01/15/2023]
Abstract
The R100W mutation in nerve growth factor is associated with hereditary sensory autonomic neuropathy V in a Swedish family. These patients develop severe loss of perception to deep pain but with apparently normal cognitive functions. To better understand the disease mechanism, we examined a knockin mouse model of HSAN V. The homozygous mice showed significant structural deficits in intra-epidermal nerve fibers (IENFs) at birth. These mice had a total loss of pain perception at ∼2 months of age and often failed to survive to adulthood. Heterozygous mutant mice developed a progressive degeneration of small sensory fibers both behaviorally and functionally: they showed a progressive loss of IENFs starting at the age of 9 months accompanied with progressive loss of perception to painful stimuli such as noxious temperature. Quantitative analysis of lumbar 4/5 dorsal root ganglia revealed a significant reduction in small size neurons, while analysis of sciatic nerve fibers revealed the heterozygous mutant mice had no reduction in myelinated nerve fibers. Significantly, the amount of NGF secreted from mouse embryonic fibroblasts were reduced from both heterozygous and homozygous mice compared to their wild-type littermates. Interestingly, the heterozygous mice showed no apparent structural alteration in the brain: neither the anterior cingulate cortex nor the medial septum including NGF-dependent basal forebrain cholinergic neurons. Accordingly, these animals did not develop appreciable deficits in tests for brain function. Our study has thus demonstrated that the NGFR100W mutation likely affects the structure and function of peripheral sensory neurons.
Collapse
Affiliation(s)
- Wanlin Yang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Neurosciences, University of California San Diego, La Jolla, CA, USA; Department of Neurology, Zhuijiang Hospital, Southern Medical University, Guangzhou, China
| | - Kijung Sung
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Wei Xu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Maria J Rodriguez
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA; Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Andrew C Wu
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Sarai A Santos
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Savannah Fang
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Rebecca K Uber
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Stephanie X Dong
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Brandon C Guillory
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Xavier Orain
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Jordan Raus
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Corrine Jolivalt
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Nigel Calcutt
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA; Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Jianqing Ding
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengbiao Wu
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
24
|
Giuliani A, Lorenzini L, Baldassarro VA, Pannella M, Cescatti M, Fernandez M, Alastra G, Flagelli A, Villetti G, Imbimbo BP, Giardino L, Calzà L. Effects of Topical Application of CHF6467, a Mutated Form of Human Nerve Growth Factor, on Skin Wound Healing in Diabetic Mice. J Pharmacol Exp Ther 2020; 375:317-331. [PMID: 32948647 DOI: 10.1124/jpet.120.000110] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/26/2020] [Indexed: 12/16/2022] Open
Abstract
Nerve growth factor (NGF) is the protein responsible for the development and maintenance of sensory skin innervation. Given the role of appropriate innervation in skin healing, NGF has been indicated as a possible prohealing treatment in pathologic conditions characterized by nerve-ending loss, such as chronic ulcers in diabetes; however, its use as a therapeutic agent is limited by its hyperalgesic effect. We tested the effect of topical application of the nonalgogenic NGF derivative hNGFP61S/R100E in two models of skin ulcer induced in dbdb diabetic mice, investigating healing time, skin histology, reinnervation, and angiogenesis using morphologic and molecular approaches. We showed that the topical administration of CHF6467, a recombinant human NGF in which an amino acid substitution (R100E) abolished the hyperalgesic effect usually associated with NGF, accelerated skin repair in experimental wounds (full-excision and pressure-ulcer) induced in diabetic mice (dbdb). CHF6467-induced acceleration of wound healing was accompanied by increased re-epithelization, reinnervation, and revascularization as assessed by histology, immunohistochemistry, and image analysis. Bioinformatic analysis of differentially expressed genes and signaling pathways in the wound tissues showed that protein kinase B-mammalian target of rapamycin was the most regulated pathway. In spite of the transdermal absorption leading to measurable, dose-dependent increases in CHF6467 plasma levels, no systemic thermal or local mechanical hyperalgesia was observed in treated mice. When tested in vitro in human cell lines, CHF6467 stimulated keratinocyte and fibroblast proliferation and tube formation by endothelial cells. Collectively, these results support a possible use of CHF6467 as a prohealing agent in skin lesions in diabetes. SIGNIFICANCE STATEMENT: Topical application of CHF6467 accelerates reinnervation, neoangiogenesis, and wound healing in diabetic mice in both full-thickness skin-excision and pressure-ulcer models through the protein kinase B/mammalian target of rapamycin pathway and does not induce hyperalgesia.
Collapse
Affiliation(s)
- A Giuliani
- Department of Veterinary Medical Science, University of Bologna, Italy (A.G., L.L., M.F., L.G.); Interdepartmental Center for Industrial Research in Life Sciences and Technologies University of Bologna, Italy (L.L., V.A.B., G.A., A.F, L.G., L.C.); Department of of Pharmacy and Biotechnology, University of Bologna, Italy (L.C.); Fondazione IRET, Ozzano Emilia, Italy (M.P., M.C.); Chiesi Farmaceutici, Parma, Italy (G.V., B.P.I.)
| | - L Lorenzini
- Department of Veterinary Medical Science, University of Bologna, Italy (A.G., L.L., M.F., L.G.); Interdepartmental Center for Industrial Research in Life Sciences and Technologies University of Bologna, Italy (L.L., V.A.B., G.A., A.F, L.G., L.C.); Department of of Pharmacy and Biotechnology, University of Bologna, Italy (L.C.); Fondazione IRET, Ozzano Emilia, Italy (M.P., M.C.); Chiesi Farmaceutici, Parma, Italy (G.V., B.P.I.)
| | - V A Baldassarro
- Department of Veterinary Medical Science, University of Bologna, Italy (A.G., L.L., M.F., L.G.); Interdepartmental Center for Industrial Research in Life Sciences and Technologies University of Bologna, Italy (L.L., V.A.B., G.A., A.F, L.G., L.C.); Department of of Pharmacy and Biotechnology, University of Bologna, Italy (L.C.); Fondazione IRET, Ozzano Emilia, Italy (M.P., M.C.); Chiesi Farmaceutici, Parma, Italy (G.V., B.P.I.)
| | - M Pannella
- Department of Veterinary Medical Science, University of Bologna, Italy (A.G., L.L., M.F., L.G.); Interdepartmental Center for Industrial Research in Life Sciences and Technologies University of Bologna, Italy (L.L., V.A.B., G.A., A.F, L.G., L.C.); Department of of Pharmacy and Biotechnology, University of Bologna, Italy (L.C.); Fondazione IRET, Ozzano Emilia, Italy (M.P., M.C.); Chiesi Farmaceutici, Parma, Italy (G.V., B.P.I.)
| | - M Cescatti
- Department of Veterinary Medical Science, University of Bologna, Italy (A.G., L.L., M.F., L.G.); Interdepartmental Center for Industrial Research in Life Sciences and Technologies University of Bologna, Italy (L.L., V.A.B., G.A., A.F, L.G., L.C.); Department of of Pharmacy and Biotechnology, University of Bologna, Italy (L.C.); Fondazione IRET, Ozzano Emilia, Italy (M.P., M.C.); Chiesi Farmaceutici, Parma, Italy (G.V., B.P.I.)
| | - M Fernandez
- Department of Veterinary Medical Science, University of Bologna, Italy (A.G., L.L., M.F., L.G.); Interdepartmental Center for Industrial Research in Life Sciences and Technologies University of Bologna, Italy (L.L., V.A.B., G.A., A.F, L.G., L.C.); Department of of Pharmacy and Biotechnology, University of Bologna, Italy (L.C.); Fondazione IRET, Ozzano Emilia, Italy (M.P., M.C.); Chiesi Farmaceutici, Parma, Italy (G.V., B.P.I.)
| | - G Alastra
- Department of Veterinary Medical Science, University of Bologna, Italy (A.G., L.L., M.F., L.G.); Interdepartmental Center for Industrial Research in Life Sciences and Technologies University of Bologna, Italy (L.L., V.A.B., G.A., A.F, L.G., L.C.); Department of of Pharmacy and Biotechnology, University of Bologna, Italy (L.C.); Fondazione IRET, Ozzano Emilia, Italy (M.P., M.C.); Chiesi Farmaceutici, Parma, Italy (G.V., B.P.I.)
| | - A Flagelli
- Department of Veterinary Medical Science, University of Bologna, Italy (A.G., L.L., M.F., L.G.); Interdepartmental Center for Industrial Research in Life Sciences and Technologies University of Bologna, Italy (L.L., V.A.B., G.A., A.F, L.G., L.C.); Department of of Pharmacy and Biotechnology, University of Bologna, Italy (L.C.); Fondazione IRET, Ozzano Emilia, Italy (M.P., M.C.); Chiesi Farmaceutici, Parma, Italy (G.V., B.P.I.)
| | - G Villetti
- Department of Veterinary Medical Science, University of Bologna, Italy (A.G., L.L., M.F., L.G.); Interdepartmental Center for Industrial Research in Life Sciences and Technologies University of Bologna, Italy (L.L., V.A.B., G.A., A.F, L.G., L.C.); Department of of Pharmacy and Biotechnology, University of Bologna, Italy (L.C.); Fondazione IRET, Ozzano Emilia, Italy (M.P., M.C.); Chiesi Farmaceutici, Parma, Italy (G.V., B.P.I.)
| | - B P Imbimbo
- Department of Veterinary Medical Science, University of Bologna, Italy (A.G., L.L., M.F., L.G.); Interdepartmental Center for Industrial Research in Life Sciences and Technologies University of Bologna, Italy (L.L., V.A.B., G.A., A.F, L.G., L.C.); Department of of Pharmacy and Biotechnology, University of Bologna, Italy (L.C.); Fondazione IRET, Ozzano Emilia, Italy (M.P., M.C.); Chiesi Farmaceutici, Parma, Italy (G.V., B.P.I.)
| | - L Giardino
- Department of Veterinary Medical Science, University of Bologna, Italy (A.G., L.L., M.F., L.G.); Interdepartmental Center for Industrial Research in Life Sciences and Technologies University of Bologna, Italy (L.L., V.A.B., G.A., A.F, L.G., L.C.); Department of of Pharmacy and Biotechnology, University of Bologna, Italy (L.C.); Fondazione IRET, Ozzano Emilia, Italy (M.P., M.C.); Chiesi Farmaceutici, Parma, Italy (G.V., B.P.I.)
| | - L Calzà
- Department of Veterinary Medical Science, University of Bologna, Italy (A.G., L.L., M.F., L.G.); Interdepartmental Center for Industrial Research in Life Sciences and Technologies University of Bologna, Italy (L.L., V.A.B., G.A., A.F, L.G., L.C.); Department of of Pharmacy and Biotechnology, University of Bologna, Italy (L.C.); Fondazione IRET, Ozzano Emilia, Italy (M.P., M.C.); Chiesi Farmaceutici, Parma, Italy (G.V., B.P.I.)
| |
Collapse
|
25
|
Ali AM, Kunugi H. Royal Jelly as an Intelligent Anti-Aging Agent-A Focus on Cognitive Aging and Alzheimer's Disease: A Review. Antioxidants (Basel) 2020; 9:E937. [PMID: 33003559 PMCID: PMC7601550 DOI: 10.3390/antiox9100937] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 02/08/2023] Open
Abstract
The astronomical increase of the world's aged population is associated with the increased prevalence of neurodegenerative diseases, heightened disability, and extremely high costs of care. Alzheimer's Disease (AD) is a widespread, age-related, multifactorial neurodegenerative disease that has enormous social and financial drawbacks worldwide. The unsatisfactory outcomes of available AD pharmacotherapy necessitate the search for alternative natural resources that can target various the underlying mechanisms of AD pathology and reduce disease occurrence and/or progression. Royal jelly (RJ) is the main food of bee queens; it contributes to their fertility, long lifespan, and memory performance. It represents a potent nutraceutical with various pharmacological properties, and has been used in a number of preclinical studies to target AD and age-related cognitive deterioration. To understand the mechanisms through which RJ affects cognitive performance both in natural aging and AD, we reviewed the literature, elaborating on the metabolic, molecular, and cellular mechanisms that mediate its anti-AD effects. Preclinical findings revealed that RJ acts as a multidomain cognitive enhancer that can restore cognitive performance in aged and AD models. It promotes brain cell survival and function by targeting multiple adversities in the neuronal microenvironment such as inflammation, oxidative stress, mitochondrial alterations, impaired proteostasis, amyloid-β toxicity, Ca excitotoxicity, and bioenergetic challenges. Human trials using RJ in AD are limited in quantity and quality. Here, the limitations of RJ-based treatment strategies are discussed, and directions for future studies examining the effect of RJ in cognitively impaired subjects are noted.
Collapse
Affiliation(s)
- Amira Mohammed Ali
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-0031, Japan;
- Department of Psychiatric Nursing and Mental Health, Faculty of Nursing, Alexandria University, Alexandria 21527, Egypt
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-0031, Japan;
- Department of Psychiatry, Teikyo University School of Medicine, Tokyo 173-8605, Japan
| |
Collapse
|
26
|
Gavrilova SI, Alvarez A. Cerebrolysin in the therapy of mild cognitive impairment and dementia due to Alzheimer's disease: 30 years of clinical use. Med Res Rev 2020; 41:2775-2803. [PMID: 32808294 DOI: 10.1002/med.21722] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/01/2020] [Accepted: 08/05/2020] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is the most common neurocognitive disorder and a global health problem. The prevalence of AD is growing dramatically, especially in low- and middle-income countries, and will reach 131.5 million cases worldwide by 2050. Therefore, developing a disease-modifying therapy capable of delaying or even preventing the onset and progression of AD has become a world priority, and is an unmet need. The pathogenesis of AD, considered as the result of an imbalance between resilience and risk factors, begins many years before the typical clinical picture develops and involves multiple pathophysiological mechanisms. Since the pathophysiology of AD is multifactorial, it is not surprising that all attempts done to modify the disease course with drugs directed towards a single therapeutic target have been unsuccessful. Thus, combined modality therapy, using multiple drugs with a single mechanism of action or multi-target drugs, appears as the most promising strategy for both effective AD therapy and prevention. Cerebrolysin, acting as a multitarget peptidergic drug with a neurotrophic mode of action, exerts long-lasting therapeutic effects on AD that could reflect its potential utility for disease modification. Clinical trials demonstrated that Cerebrolysin is safe and efficacious in the treatment of AD, and may enhance and prolong the efficacy of cholinergic drugs, particularly in moderate to advanced AD patients. In this review, we summarize advances of therapeutic relevance in the pathogenesis and the biomarkers of AD, paying special attention to neurotrophic factors, and present results of preclinical and clinical investigations with Cerebrolysin in AD.
Collapse
Affiliation(s)
- Svetlana I Gavrilova
- Department of Geriatric Psychiatry, Cognitive Disorders and Alzheimer's Disease Unit, Mental Health Research Center, Moscow, Russia
| | - Anton Alvarez
- Department of Neuropsychiatry, Medinova Institute of Neurosciences, Clinica RehaSalud, A Coruña, Spain.,Clinical Research Department, QPS Holdings, A Coruña, Spain
| |
Collapse
|
27
|
Zhou H, Gong Y, Liu Y, Huang A, Zhu X, Liu J, Yuan G, Zhang L, Wei JA, Liu J. Intelligently thermoresponsive flower-like hollow nano-ruthenium system for sustained release of nerve growth factor to inhibit hyperphosphorylation of tau and neuronal damage for the treatment of Alzheimer's disease. Biomaterials 2020; 237:119822. [DOI: 10.1016/j.biomaterials.2020.119822] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 11/26/2019] [Accepted: 01/23/2020] [Indexed: 12/19/2022]
|
28
|
Garcia-Garcia RM, Arias-Alvarez M, Sanchez-Rodriguez A, Lorenzo PL, Rebollar PG. Role of nerve growth factor in the reproductive physiology of female rabbits: A review. Theriogenology 2020; 150:321-328. [PMID: 32088037 DOI: 10.1016/j.theriogenology.2020.01.070] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 01/29/2020] [Indexed: 12/27/2022]
Abstract
Rabbit does are reflex ovulators such that coitus is needed to release GnRH and elicit the LH surge that triggers the ovulation of mature oocytes. However, the mechanisms eliciting ovulation in this species remain unclear. One of the most promising recently discovered candidates with a role in female reproductive physiology is nerve growth factor beta (β-NGF). This neurotrophin and its high-affinity receptor TrkA and low affinity receptor p75, is present in all compartments of the ovary, oviduct and uterus suggesting a physiologic role in ovarian folliculogenesis, steroidogenesis, ovulation, luteogenesis and embryo development. Besides, evidence exists that β-NGF found in seminal plasma could exert a modulatory role in the female hypothalamus-pituitary-ovarian axis contributing to the adrenergic and cholinergic neuronal stimulus of GnRH neurons in an endocrine manner during natural mating. Probably, the paracrine and local roles of the neurotrophin in steroidogenesis and ovulation reinforce the neuroendocrine pathway that leads to ovulation. This review updates knowledge of the role of β-NGF in rabbit reproduction, including its possible contribution to the mechanisms of action that induce ovulation, and discusses perspectives for the future applications of this neurotrophin on rabbit farms.
Collapse
Affiliation(s)
- R M Garcia-Garcia
- Dept. Physiology, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain.
| | - M Arias-Alvarez
- Dept. Animal Production, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
| | - A Sanchez-Rodriguez
- Dept. Physiology, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
| | - P L Lorenzo
- Dept. Physiology, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
| | - P G Rebollar
- Dept. of Agrarian Production, ETSIAAB, Polytechnic University of Madrid, Madrid, Spain
| |
Collapse
|
29
|
The NGF R100W Mutation Specifically Impairs Nociception without Affecting Cognitive Performance in a Mouse Model of Hereditary Sensory and Autonomic Neuropathy Type V. J Neurosci 2019; 39:9702-9715. [PMID: 31685654 DOI: 10.1523/jneurosci.0688-19.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 10/01/2019] [Accepted: 10/27/2019] [Indexed: 12/22/2022] Open
Abstract
Nerve growth factor (NGF) is a key mediator of nociception, acting during the development and differentiation of dorsal root ganglion (DRG) neurons, and on adult DRG neuron sensitization to painful stimuli. NGF also has central actions in the brain, where it regulates the phenotypic maintenance of cholinergic neurons. The physiological function of NGF as a pain mediator is altered in patients with Hereditary Sensory and Autonomic Neuropathy type V (HSAN V), caused by the 661C>T transition in the Ngf gene, resulting in the R100W missense mutation in mature NGF. Homozygous HSAN V patients present with congenital pain insensitivity, but are cognitively normal. This led us to hypothesize that the R100W mutation may differentially affect the central and peripheral actions of NGF. To test this hypothesis and provide a mechanistic basis to the HSAN V phenotype, we generated transgenic mice harboring the human 661C>T mutation in the Ngf gene and studied both males and females. We demonstrate that heterozygous NGFR100W/wt mice display impaired nociception. DRG neurons of NGFR100W/wt mice are morphologically normal, with no alteration in the different DRG subpopulations, whereas skin innervation is reduced. The NGFR100W protein has reduced capability to activate pain-specific signaling, paralleling its reduced ability to induce mechanical allodynia. Surprisingly, however, NGFR100W/wt mice, unlike heterozygous mNGF+/- mice, show no learning or memory deficits, despite a reduction in secretion and brain levels of NGF. The results exclude haploinsufficiency of NGF as a mechanistic cause for heterozygous HSAN V mice and demonstrate a specific effect of the R100W mutation on nociception.SIGNIFICANCE STATEMENT The R100W mutation in nerve growth factor (NGF) causes Hereditary Sensory and Autonomic Neuropathy type V, a rare disease characterized by impaired nociception, even in apparently clinically silent heterozygotes. For the first time, we generated and characterized heterozygous knock-in mice carrying the human R100W-mutated allele (NGFR100W/wt). Mutant mice have normal nociceptor populations, which, however, display decreased activation of pain transduction pathways. NGFR100W interferes with peripheral and central NGF bioavailability, but this does not impact on CNS function, as demonstrated by normal learning and memory, in contrast with heterozygous NGF knock-out mice. Thus, a point mutation allows neurotrophic and pronociceptive functions of NGF to be split, with interesting implications for the treatment of chronic pain.
Collapse
|
30
|
Valdovinos-Flores C, Limón-Pacheco JH, León-Rodríguez R, Petrosyan P, Garza-Lombó C, Gonsebatt ME. Systemic L-Buthionine -S-R-Sulfoximine Treatment Increases Plasma NGF and Upregulates L-cys/L-cys2 Transporter and γ-Glutamylcysteine Ligase mRNAs Through the NGF/TrkA/Akt/Nrf2 Pathway in the Striatum. Front Cell Neurosci 2019; 13:325. [PMID: 31396052 PMCID: PMC6664075 DOI: 10.3389/fncel.2019.00325] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/03/2019] [Indexed: 01/31/2023] Open
Abstract
Glutathione (GSH) is the most abundant intracellular antioxidant. GSH depletion leads to oxidative stress and neuronal damage in the central nervous system (CNS). In mice, the acute systemic inhibition of GSH synthesis by L-buthionine-S-R-sulfoximine (BSO) triggers a protective response and a subsequent increase in the CNS GSH content. This response might be modulated by a peripheral increment of circulating nerve growth factor (NGF). NGF is an important activator of antioxidant pathways mediated by tropomyosin-related kinase receptor A (TrkA). Here, we report that peripheral administration of BSO increased plasma NGF levels. Additionally, BSO increased NGF levels and activated the NGF/TrkA/Akt pathway in striatal neurons. Moreover, the response in the striatum included an increased transcription of nrf2, gclm, lat1, eaac1, and xct, all of which are involved in antioxidant responses, and L-cys/L-cys2 and glutamate transporters. Using antibody against NGF confirmed that peripheral NGF activated the NGF/TrkA/Akt/Nrf2 pathway in the striatum and subsequently increased the transcription of gclm, nrf2, lat1, eaac1, and xct. These results provide evidence that the reduction of peripheral GSH pools increases peripheral NGF circulation that orchestrates a neuroprotective response in the CNS, at least in the striatum, through the NGF/TrkA/Akt/Nrf2 pathway.
Collapse
Affiliation(s)
- Cesar Valdovinos-Flores
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Jorge H Limón-Pacheco
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Renato León-Rodríguez
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Pavel Petrosyan
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Carla Garza-Lombó
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Maria E Gonsebatt
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
31
|
Hefti F. Pharmacology of nerve growth factor and discovery of tanezumab, an anti-nerve growth factor antibody and pain therapeutic. Pharmacol Res 2019; 154:104240. [PMID: 31026504 DOI: 10.1016/j.phrs.2019.04.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 04/19/2019] [Indexed: 01/01/2023]
Abstract
The pharmacology of Nerve Growth Factor (NGF) and the discovery and development of tanezumab, a monoclonal anti-NGF antibody for the treatment of pain illustrate the complex and unpredictable nature of modern drug development. Initial efforts attempted to use NGF agonistically for Alzheimer's disease and neuropathies. Most unexpectedly, clinical studies unmasked hyperalgesic effects. These observations together with new data emerging from molecular and animal model studies stimulated the idea of using an NGF antagonist for chronic pain. These events also reflect the advances of neuropharmacology from classical small molecule efforts directed at neurotransmitter receptors to modern biotechnology with significant integration in molecular biology, biochemistry, and protein engineering.
Collapse
Affiliation(s)
- Franz Hefti
- Prevail Therapeutics Inc., 430 East 29th Street, New York, NY, 10016, USA.
| |
Collapse
|
32
|
Cholinergic striatal neurons are increased in HSAN V homozygous mice despite reduced NGF bioavailability. Biochem Biophys Res Commun 2019; 509:763-766. [DOI: 10.1016/j.bbrc.2018.12.178] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 12/28/2018] [Indexed: 12/11/2022]
|
33
|
Alzheimer's disease: understanding homeostasis deregulation to foster development of effective therapies. Pharmacol Res 2018; 139:467-468. [PMID: 30553822 DOI: 10.1016/j.phrs.2018.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|