Abstract
Since publishing our earlier report describing a strategy for the treatment of central nervous system (CNS) diseases by inhibiting the cell cycle and without disrupting neurogenesis (Liu et al. 2010), we now update and extend this strategy to applications in the treatment of cancers as well. Here, we put forth the concept of “aberrant cell cycle diseases” to include both cancer and CNS diseases, the two unrelated disease types on the surface, by focusing on a common mechanism in each aberrant cell cycle reentry. In this paper, we also summarize the pharmacological approaches that interfere with classical cell cycle molecules and mitogenic pathways to block the cell cycle of tumor cells (in treatment of cancer) as well as to block the cell cycle of neurons (in treatment of CNS diseases). Since cell cycle inhibition can also block proliferation of neural progenitor cells (NPCs) and thus impair brain neurogenesis leading to cognitive deficits, we propose that future strategies aimed at cell cycle inhibition in treatment of aberrant cell cycle diseases (i.e., cancers or CNS diseases) should be designed with consideration of the important side effects on normal neurogenesis and cognition.
1. Introduction
The cell cycle is an irreversible, ordered set of events that normally leads to cellular division [1–5]. The release of cells from a quiescent state (G0) results in their entry into the first gap phase (G1), during which the cells prepare for DNA replication in the synthetic phase (S). This is followed by the second gap phase (G2) and mitosis phase (M). After the cell has split into its two daughter cells, the new cells enter either G1 or G0. Tumors usually originate from adult tissues, in which the majority of cells are in the G0 quiescent phase [4]. Mature neurons normally maintain themselves in G0 resting phase. These facts suggest that the cells that go on to form tumors and mature neurons share a common G0 state of quiescence.
Since cell cycle is irreversible, this raises a possibility that irreversible cell cycle reentry mediates the irreversible neuronal death that mirrors the irreversible progression of some central nervous system (CNS) diseases, such as Alzheimer's disease (AD). If this is true, it will partially explain why AD is incurable once even early AD symptoms occur, for the early AD symptoms may indicate that the neurons have reentered the cell cycle that ends up leading to neuronal death and AD progression. Thus, the best strategy in treatment of CNS diseases is to prevent cell cycle re-entry at the early stage before neurons leave the G0 phase at all, since even the mere entrance into the initial cell cycle may lead to unavoidable neuronal death.
Since re-entry into the cell cycle by tumor cells or neurons has been associated with many tumor or CNS diseases and linked to uncontrolled cell proliferation (in cancer) or neuronal death (in CNS diseases), cell cycle inhibition strategies are of interest in the treatment of both tumor and CNS diseases. For instance, the cell cycle inhibitors, such as cyclin-dependent kinase (CDK) inhibitors, have been widely studied as cancer therapeutics. They have been used to inhibit growth of several types of tumor cells in numerous preclinical studies, both in vitro and in vivo [6–12]. Several cell cycle inhibitors have advanced to human clinical trials for evaluation as a treatment for a broad range of solid tumors and hematological malignancies such as chronic lymphocytic leukemia (CLL) [13–17]. Though no clinical trials of the cell cycle inhibitors are reported in the treatment of CNS diseases, preclinical experiments demonstrate that the cell cycle inhibitors improve behavioral outcomes and increase neuronal survival in a series of CNS disease models [18–33].
Cell cycle inhibition kills tumor cells (in treatment of cancer) or protects mature neurons from death (in treatment of CNS diseases), whereas this can also block proliferation of neural progenitor cells (NPCs) and thus impair brain neurogenesis leading to cognitive deficits in the patients of cancer and CNS diseases [1]. Since the presence of cognitive deficits is a major factor markedly affecting quality of life of these patients, the cell cycle inhibition strategy in treatment of cancer and CNS diseases should consider the consequences on other cell types that can be affected, such as NPCs.
As a way to describe the two seemingly different disease types (i.e., cancer and CNS diseases) that share the common mechanism of cell cycle re-entry, we propose a broader term of “aberrant cell cycle diseases”—one which includes not only cancers but also CNS diseases. A detailed description of how the cell cycle re-entry, at least in part, underlies cancers and CNS diseases follows before we discuss the pharmacological approaches that have been examined in therapeutic treatment of the two disease types.
2. Aberrant Cell Cycle Diseases: Cancers and CNS Diseases
Cancers and CNS diseases are two major threats to human health. Epidemiological studies show that patients with CNS disease, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and multiple sclerosis (MS), have a significantly lower risk of most cancers [34–38]. The reverse correlations also hold true: cancer survivors have a significantly lower risk of developing some of these CNS diseases. However, there are exceptions: Parkinson's patients have an increased risk for melanoma [39–44], autism patients have an increased risk for breast cancer [45], and Fragile X Syndrome patients have increased risk for lip cancer [46]. No matter what associations underlie certain cancers and CNS diseases, these correlative studies have raised an interesting question: what associated processes or mechanisms do dying neurons and growing tumor cells have in common?
Aberrant cell cycle is the hallmark of many tumor cells in cancers [47–49] and is also observed in postmortem and/or animal studies of dying neurons in a series of CNS diseases, such as AD, PD, stroke, epilepsy, cerebral hypoxia-ischemia, amyotrophic lateral sclerosis (ALS), traumatic brain injury (TBI), among others [18–33, 50–64]. Although tumor cells undergo uncontrolled proliferation, many tumors originate from adult tissues in which the majority of cells are in the G0 quiescent phase [4]. Similarly, mature neurons stay in G0 quiescent phase in normal physiological conditions, but do reenter the cell cycle irregularly (and die) in certain pathological conditions.
Thus, the cells that go on to form tumors and healthy neurons share a common G0 state of quiescence. However, if tumor cells re-enter the cell cycle, they survive and often proliferate, whereas mature neurons will die. Therefore, cell cycle inhibition can be applied not only to kill tumor cells (in treatment of cancer), but also to protect neurons from death (in treatment of CNS diseases). This is strongly supported by the findings that mutation of PARK2, a tumor suppressor gene, results not only in neuronal death in Parkinson's disease, but also in tumor cell proliferation in glioblastoma and other human cancer [65]. Consistently, cell cycle inhibition mot only promotes the death of naive PC12 (pheochromocytoma) tumor cells, but also prevents the death of nerve growth factor- (NGF-) differentiated PC12 neuronal cells [66, 67].
3. Cell Cycle and “Expanded Cell Cycle”
Classically, the proteins and regulators of the cell cycle include CDKs, cyclins, CDK inhibitors, and CDK substrates. However, the cell cycle inhibition strategies that target on these molecules lack specificity for their target tumor cells or dying neurons and thus interfere the normal biological processes performed by other cell types, since the core cell-cycle-associated molecules are often highly conserved throughout eukaryotes, thus we proposed an “expanded cell cycle” to include not only the core cell cycle molecules mentioned above, but also the mitogenic molecules and the signaling pathways that interact with them. Under specific environmental and/or pathological conditions, such as exposure of tobacco smoke, benzene, ultraviolet B radiation, and/or enhancement of mitogenic molecules (i.e., thrombin, growth factors, amyloid beta, etc.), activation of specific pathways to mediate abnormal cell cycle re-entry may arise and thus trigger tumorigenesis of normal cells and/or death of neurons. It is always impossible to prove exactly what caused a cancer or CNS disease in any individual, because most of these diseases have multivariate causes. However, the various causes seem to result in a common outcome—cell cycle re-entry, mediated by several common mitogenic pathways. The main mitogenic pathways include (1) FAK/Src/Ras/Raf/MEK1, 2/ERK1, 2 → cell cycle re-entry [68–72]; (2) Ras/Rac1/MEK3, 6/P38 → cell cycle re-entry [73, 74]; (3) PLC/IP3/PKC/JNK → cell cycle re-entry [75, 76]; (4) PI3K/Akt/mTOR/Tau → cell cycle re-entry [60, 77, 78]; (5) JAK/STAT → cell cycle re-entry [79, 80]. In addition, many molecules, including ROS, PGE2, NO, and Ca2+, can directly or indirectly participate in the main mitogenic signaling pathways [81–86]. The idea of an “expanded cell cycle” provides a wider view encompassing a broad range of molecules representing potential targets and thus approaches that can serve as treatments for cancer and CNS diseases—all sharing the common outcome of cell cycle inhibition.
4. Pharmacological Approaches Interfering with the “Expanded Cell Cycle” in Treatment of Aberrant Cell Cycle Diseases
In theory any part of the “expanded cell cycle” could act as a potential target for drug discovery. For example, thrombin activation is substantially increased in cancers [87–89] and CNS diseases (i.e., AD, stroke). Thrombin may then go on to activate Src kinases [90, 91]. Src kinases will activate MAPK which will activate CDK4/cyclinD complexes and promote cell cycle re-entry [71, 91, 92]. Thus, these molecules (thrombin, Src kinases, and MAPK), while not considered traditional components of the cell cycle, would all be considered part of the “expanded cell cycle.” Similarly, other protein kinases (including JAK, Akt, PKC, JNK, ERK, GSK-3β.) are also important molecules in the mitogenic pathways leading to neuronal cell cycle re-entry. Most targets mentioned above have been examined in cancer therapies as well as in CNS diseases. Pharmacological approaches based on those targets include traditional cell cycle inhibitors (CDK inhibitors), antioxidants, NMDA-receptor modulators, i/eNOS inhibitors, COX-2 inhibitors, protein kinase inhibitors, and others (Table 1).
Table 1.
Treatments | ||||
---|---|---|---|---|
Targets | Cancers | CNS diseases | ||
Agents | Stages | Agents | Stages | |
CDK inhibitors | Flavopiridol, Indisulam, AZD5438 SNS-032 Bryostatin-1 Seliciclib PD 0332991 SCH 727965 UCN-01, Roscovitine, AT7519 |
Clinical trials in a broad range of solid tumors and chronic lymphocytic leukemia (CLL) [13–16]. | Flavopiridol Olomoucine Roscovitine Quinazolines |
Experimental trials in AD [18–22], PD [21], stroke [23, 24], TBI [25, 26], SCI [27, 28], excitotoxic stress [29–32], optic nerve transaction [33]. |
| ||||
Antioxidants | Isoliquiritigenin | Experimental trials in prostate cancer [124]. | Edaravone NXY-059 Coenzyme Q10 Vitamin E Melatonin Trolox SOD NAC PBN |
Clinical use in stroke in Asia [125, 126], Clinical trials in stroke [126], AD [127–129], ALS [130–132]. Experimental trials in ALS [133], PD [134], SCI [135]. |
| ||||
i/eNOS inhibitors | L-NAME AMT NPA |
Experimental trials in prostate cancer [136]. | L-NAME AMT NPA |
Experimental trials in ICH [137], SCI [135]. |
| ||||
Cox-2 inhibitors | Celecoxib Rofecoxib NS-398 |
Clinical trials in bladder cancer [138], lung cancer [139, 140], head and neck cancer [141], pancreatic cancer [142], prostate cancer [143], breast cancer [144], colorectal cancer [145]. Experimental trials in pancreatic cancer [146], prostate cancer [147], esophageal cancer [148], colon cancer [149]. |
Celecoxib Rofecoxib NS-398 |
Clinical trials in AD [150, 151]. Experimental trials in ICH [137]. |
| ||||
Ca2+ channel blockers | KYS05090 | Experimental trials in cancers [152]. | Flunarizine | Clinical trials in stroke [153]. |
| ||||
Glutamatergic modulators | MK-801 | Experimental trials in breast cancer [154], brain tumors [155]. | Riluzole Ceftriaxone Talampanel MK-801 NBQX |
Clinical trials in ALS [156]. Experimental trials in TBI [157, 158], SCI [159], ICH [160], stroke [161]. |
| ||||
NMDA-receptor modulators | AP5 Memantine | Experimental trials in breast cancer [154], gastric cancer [162]. | Memantine | Clinical use in AD [163–166]. Clinical trials in ALS [156]. Off-label use in psychiatric disorders [167]. Experimental trials in TBI [168], ICH [169], stroke [170], |
| ||||
Thrombin inhibitors | Heparin | Experimental trials in lung cancer [171]. | Heparin Hirudin |
Experimental trials in ischemic stroke [172], ICH [118, 173, 174]. |
| ||||
Thrombin receptor-1 antagonist | RWJ-58259 SCH-79797 |
Experimental trials in colon cancer [175]. | BMS-200261 | Experimental trials in stroke [176]; PD [177]. |
| ||||
Ras inhibitors | Lovastatin FTS |
Clinical trials in neurofibroma [178], head and neck cancer [179]. Experimental trials in neurofibroma [180], liver cancer [181], ovarian cancer [182], breast cancer [183], prostate cancer [184], lung cancer [185]. |
Lovastatin Exoenzyme |
Clinical trials in acute ischemic stroke [186, 187]. Experimental trials in motor neuron disorders [188, 189]. |
| ||||
Src Inhibitors | KX-01 Dasatinib PP1 PP2 Saracatinib |
Clinical trials in breast cancer [190–192]. Experimental trials in breast cancer [193], lung cancer [194, 195], cervical cancer [196], renal cancer [197], prostate cancer [198]. |
PP1 PP2 |
Experimental trials in ICH [91, 199], AD [70]. |
| ||||
JAK/Stat Inhibitors | EGCG WP-1034 |
Experimental trials in prostate cancer [200], breast cancer [201], lung cancer [202], pancreatic cancer [203], acute myeloid leukemia [204]. | EGCG | Experimental trials in AD, PD, HIV associated Dementia, multiple sclerosis (MS), ALS, or Pick's Disease [79]. |
| ||||
GSK-3β inhibitors | Lithium SB 415286 SB 216763 AR-A014418 |
Experimental trials in colon cancer [205], neuroblastoma [206]. | L803-mt Lithium Kenpaullone Indirubin SB 216763 SB 415286 |
Experimental trials in AD [207–212], PD [213], brain injury [214]. Clinical trials in ALS [156, 215]. |
| ||||
PI3K inhibitors | NVP-BEZ235 GSK2126458 |
Experimental trials in breast cancer [216]. | LY 294002 | Experimental trials in AD [217], PD [218]. |
| ||||
Akt Inhibitors | Perifosine MK-2206 RX-0201 PBI-05204 GSK2141795 |
Clinical trials in advanced cancer [219, 220]. | LY 294002 | Experimental trials in AD [217], PD [218]. |
| ||||
m-TOR | Everolimus Temsirolimus |
Clinical use in pancreatic cancer [221], renal cancer [221]. Clinical trials in epithelial ovarian cancer/primary peritoneal cancer [222], Endometrial Cancer [223], glioblastoma [224], breast cancer [225], neuroendocrine tumours [226], lung cancer [227], bladder cancer [228], renal cancer [229, 230]. |
Rapamycin | Experimental trials in AD [231], PD [232], TBI [233, 234], SCI [235, 236], stroke [237]. |
| ||||
Tau inhibitor | SG410 | Experimental trials in pancreas cancer [238]. | TRx-0014 | Clinical use in AD [239, 240]. |
| ||||
ERK1/2 kinase pathway | PD98059 | Experimental trials in prostate cancer [241], lung cancer [242]. | PD98059 | Experimental trials in ICH [72, 92]. |
| ||||
P38 kinase pathway | SB203580 | Experimental trials in colon cancer [243]. | SB203580 SB239063 |
Experimental trials in ICH [92], PD [244], stroke [245]. |
| ||||
JNK kinase pathway | SP600125 | Experimental trials in cancer cells [246, 247]. | CEP-1347 Colostrinin SP600125 |
Experimental trials in ICH [72, 92], AD [248, 249], stroke [250], PD [248, 250]. |
4-Amino-5-(4-methylphenyl)-7-(t-butyl)pyrazolo(3,4-d)pyrimidine (PP1), 4-Amino-5-(4-chlorophenyl)-7(t-butyl)pyrazol(3,4-d)pyramide (PP2), 2-Amino-5,6-dihydro-6-methyl-4H-1,3-thiazine hydrochloride (AMT), 2,3-Dihydroxy-6-nitro-7-sulfamoyl-benzo [f]quinoxaline-2,3-dione (NBQX), Dl-2-amino-5-phosphonovaleric acid (AP5), Epigallocatechin-3-gallate (EGCG), Farnesylthiosalicylic acid (FTS), Methylthioinnium chloride (TRx-0014), 5-Methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine (MK 801), N-acytyl-L-cysteine (NAC), N(G)-nitro-l-arginine methyl ester (l-NAME), N(omega)-propyl-l-arginine (NPA), Phenyl-N-tert-butylnitrone (PBN), Superoxide dismutase (SOD).
5. Cognitive Side Effects of Clinic Therapies for Aberrant Cell Cycle Diseases
In treatment of peripheral or brain cancers, surgical removal of the tumor is recommended whenever possible. Anticancer medications (chemotherapy, CT) may be prescribed, as well as radiation therapy (RT). CT- or RT is cytotoxic, not only slowing down or killing rapidly dividing cells and producing many different types of DNA damage [93], but detrimentally leading to damage of normal tissue, especially of fast-growing healthy cells, including neural progenitor cells (NPCs), and also red and white blood cells [94, 95]. Recent reports show that CT results in cognitive side effects in extracranial cancer (i.e., breast cancer, prostate, etc.) patients [96–98], and CT or RT has also been associated with neurogenesis impairment and long-term cognitive deficits in brain cancer patients [99–105]. The main idea that is thought to contribute to the CT or RT-induced cognitive decline is that these treatments block proliferation of NPCs in the hippocampal and periventricular zones, which cannot be repopulated as healthy cells die. This leads to cognitive decline, since the NPCs help maintain neurogenesis [106–109], repair damage from brain injury [105, 109–118], and are important in cognition [119–121].
Besides the CT and RT mentioned above in treatment of cancers, some pharmacological approaches that interfere with classical cell cycle molecules or mitogenic pathways have been examined in cancer and CNS disease therapies (Table 1). Therapies directed at any component inhibiting the cell cycle must be as specific as possible considering cell cycle re-entry contributes to the proliferation of tumor cells, the death of mature neurons, and the genesis of NPCs in adult brain. Therefore, any therapeutics that prevent tumor growth and/or neuronal death by blocking cell cycle re-entry may have limited benefit because they may impair neurogenesis and thus lead to cognitive side effects. This may provide at least a partial explanation for the questionable efficacy of some currently approved drugs, such as the NMDA receptor modulator Memantine, in the clinical treatment of AD [122], since NMDA receptor inhibition has been shown to block progenitor cell proliferation and lead to impaired neurogenesis [123].
The cognitive side effects may be explained by the fact that current cell cycle inhibition strategies are not cell specific and also block the proliferation of important brain progenitor cells, thus impairing adult brain neurogenesis. If drugs that block the cell cycle are used to kill tumor cells (in treatment of cancer) and/or help protect neurons (in treatment of CNS diseases), it is likely that compounds would need to directly (or indirectly) block tumor and neuronal cell cycle re-entry and yet not affect the ongoing process of neurogenesis. This will only be possible if the signaling mechanisms are different in NPCs that divide in the adult brain, versus tumor cells and neurons that re-enter the cell cycle irregularly.
6. Conclusions
Cancer and CNS diseases, two seemingly different disease types, at least in part share the common molecular pathology of cell cycle re-entry. With this knowledge in mind, novel insights into cell cycle inhibition strategies to be used in treatment of the “aberrant cell cycle diseases” may be made. Future studies aimed at better understanding the respective cell cycle pathways of tumor cells, neurons, and NPCs are probably necessary before choosing the best drug targets for treating certain “aberrant cell cycle diseases” so as to consider the most effective benefits to the patient without causing indirect harm in related, but different systems.
Acknowledgment
The authors acknowledge the support of the University of California Innovative Development Award (D. Z. Liu).
References
- 1.Liu DZ, Ander BP, Sharp FR. Cell cycle inhibition without disruption of neurogenesis is a strategy for treatment of central nervous system diseases. Neurobiology of Disease. 2010;37(3):549–557. doi: 10.1016/j.nbd.2009.11.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Novak B, Tyson JJ, Gyorffy B, Csikasz-Nagy A. Irreversible cell-cycle transitions are due to systems-level feedback. Nature Cell Biology. 2007;9(7):724–728. doi: 10.1038/ncb0707-724. [DOI] [PubMed] [Google Scholar]
- 3.Norbury C, Nurse P. Animal cell cycles and their control. Annual Review of Biochemistry. 1992;61:441–470. doi: 10.1146/annurev.bi.61.070192.002301. [DOI] [PubMed] [Google Scholar]
- 4.Malumbres M, Barbacid M. To cycle or not to cycle: a critical decision in cancer. Nature Reviews Cancer. 2001;1(3):222–231. doi: 10.1038/35106065. [DOI] [PubMed] [Google Scholar]
- 5.Schwartz GK, Shah MA. Targeting the cell cycle: a new approach to cancer therapy. Journal of Clinical Oncology. 2005;23(36):9408–9421. doi: 10.1200/JCO.2005.01.5594. [DOI] [PubMed] [Google Scholar]
- 6.Erickson S, Sangfelt O, Heyman M, Castro J, Einhorn S, Grandér D. Involvement of the Ink4 proteins p16 and p15 in T-lymphocyte senescence. Oncogene. 1998;17(5):595–602. doi: 10.1038/sj.onc.1201965. [DOI] [PubMed] [Google Scholar]
- 7.Shapiro GI, Koestner DA, Matranga CB, Rollins BJ. Flavopiridol induces cell cycle arrest and p53-independent apoptosis in non-small cell lung cancer cell lines. Clinical Cancer Research. 1999;5(10):2925–2938. [PubMed] [Google Scholar]
- 8.Chien M, Astumian M, Liebowitz D, Rinker-Schaeffer C, Stadler WM. In vitro evaluation of flavopiridol, a novel cell cycle inhibitor, in bladder cancer. Cancer Chemotherapy and Pharmacology. 1999;44(1):81–87. doi: 10.1007/s002800050948. [DOI] [PubMed] [Google Scholar]
- 9.Schwartz GK, Farsi K, Maslak P, Kelsen DP, Spriggs D. Potentiation of apoptosis by flavopiridol in mitomycin-C-treated gastric and breast cancer cells. Clinical Cancer Research. 1997;3(9):1467–1472. [PubMed] [Google Scholar]
- 10.Arguello F, Alexander M, Sterry JA, et al. Flavopiridol induces apoptosis of normal lymphoid cells, causes immunosuppression, and has potent antitumor activity in vivo against human leukemia and lymphoma xenografts. Blood. 1998;91(7):2482–2490. [PubMed] [Google Scholar]
- 11.Drees M, Dengler WA, Roth T, et al. Flavopiridol (L86-8275): selective antitumor activity in vitro and activity in vivo for prostate carcinoma cells. Clinical Cancer Research. 1997;3(2):273–279. [PubMed] [Google Scholar]
- 12.Tirado OM, Mateo-Lozano S, Notario V. Roscovitine is an effective inducer of apoptosis of Ewing’s sarcoma family tumor cells in vitro and in vivo. Cancer Research. 2005;65(20):9320–9327. doi: 10.1158/0008-5472.CAN-05-1276. [DOI] [PubMed] [Google Scholar]
- 13.Wyatt PG, Woodhead AJ, Berdini V, et al. Identification of N-(4-piperidinyl)-4-(2,6-dichlorobenzoylamino)-1H- pyrazole-3-carboxamide (AT7519), a novel cyclin dependent kinase inhibitor using fragment-based X-ray crystallography and structure based drug design. Journal of Medicinal Chemistry. 2008;51(16):4986–4999. doi: 10.1021/jm800382h. [DOI] [PubMed] [Google Scholar]
- 14.Senderowicz AM. Flavopiridol: the first cyclin-dependent kinase inhibitor in human clinical trials. Investigational New Drugs. 1999;17(3):313–320. doi: 10.1023/a:1006353008903. [DOI] [PubMed] [Google Scholar]
- 15.Whittaker SR, Te Poele RH, Chan F, et al. The cyclin-dependent kinase inhibitor seliciclib (R-roscovitine; CYC202) decreases the expression of mitotic control genes and prevents entry into mitosis. Cell Cycle. 2007;6(24):3114–3131. doi: 10.4161/cc.6.24.5142. [DOI] [PubMed] [Google Scholar]
- 16.Senderowicz AM, Sausville EA. Preclinical and clinical development of cyclin-dependent kinase modulators. Journal of the National Cancer Institute. 2000;92(5):376–387. doi: 10.1093/jnci/92.5.376. [DOI] [PubMed] [Google Scholar]
- 17.Dickson MA, Schwartz GK. Development of cell-cycle inhibitors for cancer therapy. Current Oncology. 2009;16(2):36–43. doi: 10.3747/co.v16i2.428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Copani A, Ubertia D, Sortino MA, Bruno V, Nicoletti F, Memo M. Activation of cell-cycle-associated proteins in neuronal death: a mandatory or dispensable path? Trends in Neurosciences. 2001;24(1):25–31. doi: 10.1016/s0166-2236(00)01663-5. [DOI] [PubMed] [Google Scholar]
- 19.Verdaguer E, Jordá EG, Canudas AM, et al. Antiapoptotic effects of roscovitine in cerebellar granule cells deprived of serum and potassium: a cell cycle-related mechanism. Neurochemistry International. 2004;44(4):251–261. doi: 10.1016/s0197-0186(03)00147-5. [DOI] [PubMed] [Google Scholar]
- 20.Jorda EG, Verdaguer E, Canudas AM, et al. Neuroprotective action of flavopiridol, a cyclin-dependent kinase inhibitor, in colchicine-induced apoptosis. Neuropharmacology. 2003;45(5):672–683. doi: 10.1016/s0028-3908(03)00204-1. [DOI] [PubMed] [Google Scholar]
- 21.Kruman I, Schwartz E. Methods of neuroprotection by cyclin-dependent kinase inhibition. US 20080182853, 2006.
- 22.Barvian MR, Dobrusin EM, Kaltenbronn JS, et al. Quinazolines and their use for inhibiting cyclin-dependent kinase enzymes. US 6982260, 2006.
- 23.Osuga H, Osuga S, Wang F, et al. Cyclin-dependent kinases as a therapeutic target for stroke. Proceedings of the National Academy of Sciences of the United States of America. 2000;97(18):10254–10259. doi: 10.1073/pnas.170144197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Wang F, Corbett D, Osuga H, et al. Inhibition of cyclin-dependent kinases improves CA1 neuronal survival and behavioral performance after global ischemia in the rat. Journal of Cerebral Blood Flow and Metabolism. 2002;22(2):171–182. doi: 10.1097/00004647-200202000-00005. [DOI] [PubMed] [Google Scholar]
- 25.Di Giovanni S, Movsesyan V, Ahmed F, et al. Cell cycle inhibition provides neuroprotection and reduces glial proliferation and scar formation after traumatic brain injury. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(23):8333–8338. doi: 10.1073/pnas.0500989102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hilton GD, Stoica BA, Byrnes KR, Faden AI. Roscovitine reduces neuronal loss, glial activation, and neurologic deficits after brain trauma. Journal of Cerebral Blood Flow and Metabolism. 2008;28(11):1845–1859. doi: 10.1038/jcbfm.2008.75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Di Giovanni S, Knoblach SM, Brandoli C, Aden SA, Hoffman EP, Faden AI. Gene profiling in spinal cord injury shows role of cell cycle neuronal death. Annals of Neurology. 2003;53(4):454–468. doi: 10.1002/ana.10472. [DOI] [PubMed] [Google Scholar]
- 28.Tian DS, Yu ZY, Xie MJ, Bu BT, Witte OW, Wang W. Suppression of astroglial scar formation and enhanced axonal regeneration associated with functional recovery in a spinal cord injury rat model by the cell cycle inhibitor olomoucine. Journal of Neuroscience Research. 2006;84(5):1053–1063. doi: 10.1002/jnr.20999. [DOI] [PubMed] [Google Scholar]
- 29.Park DS, Obeidat A, Giovanni A, Greene LA. Cell cycle regulators in neuronal death evoked by excitotoxic stress: implications for neurodegeneration and its treatment. Neurobiology of Aging. 2000;21(6):771–781. doi: 10.1016/s0197-4580(00)00220-7. [DOI] [PubMed] [Google Scholar]
- 30.Verdaguer E, Jiménez A, Canudas AM, et al. Inhibition of cell cycle pathway by flavopiridol promotes survival of cerebellar granule cells after an excitotoxic treatment. Journal of Pharmacology and Experimental Therapeutics. 2004;308(2):609–616. doi: 10.1124/jpet.103.057497. [DOI] [PubMed] [Google Scholar]
- 31.Verdaguer E, Jordà EG, Canudas AM, et al. 3-Amino thioacridone, a selective cyclin-dependent kinase 4 inhibitor, attenuates kainic acid-induced apoptosis in neurons. Neuroscience. 2003;120(3):599–603. doi: 10.1016/s0306-4522(03)00424-x. [DOI] [PubMed] [Google Scholar]
- 32.Verdaguer E, Jordà EG, Stranges A, et al. Inhibition of CDKs: a strategy for preventing kainic acid-induced apoptosis in neurons. Annals of the New York Academy of Sciences. 2003;1010:671–674. doi: 10.1196/annals.1299.122. [DOI] [PubMed] [Google Scholar]
- 33.Lefèvre K, Clarke PGH, Danthe EE, Castagné V. Involvement of cyclin-dependent kinases in axotomy-induced retinal ganglion cell death. Journal of Comparative Neurology. 2002;447(1):72–81. doi: 10.1002/cne.10215. [DOI] [PubMed] [Google Scholar]
- 34.Roe CM, Behrens MI, Xiong C, Miller JP, Morris JC. Alzheimer disease and cancer. Neurology. 2005;64(5):895–898. doi: 10.1212/01.WNL.0000152889.94785.51. [DOI] [PubMed] [Google Scholar]
- 35.Bennett DA. Is there a link between cancer and Alzheimer disease? Neurology. 2010;75(13):1216–1217. [PubMed] [Google Scholar]
- 36.Becker C, Brobert GP, Johansson S, Jick SS, Meier CR. Cancer risk in association with Parkinson disease: a population-based study. Parkinsonism and Related Disorders. 2010;16(3):186–190. doi: 10.1016/j.parkreldis.2009.11.005. [DOI] [PubMed] [Google Scholar]
- 37.Sørensen SA, Fenger K, Olsen JH. Significantly lower incidence of cancer among patients with Huntington disease: an apoptotic effect of an expanded polyglutamine tract? Cancer. 1999;86(7):1342–1346. [PubMed] [Google Scholar]
- 38.Hofer S, Linnebank M, Weller M. Cancer risk among patients with multiple sclerosis and their parents. Neurology. 2010;74(7):614–615. doi: 10.1212/WNL.0b013e3181c777b7. [DOI] [PubMed] [Google Scholar]
- 39.Clements CM, McNally RS, Conti BJ, Mak TW, Ting JPY. DJ-1, a cancer- and Parkinson’s disease-associated protein, stabilizes the antioxidant transcriptional master regulator Nrf2. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(41):15091–15096. doi: 10.1073/pnas.0607260103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Zanetti R, Rosso S, Loria DI. Parkinson’s disease and cancer. Cancer Epidemiology Biomarkers and Prevention. 2007;16(6):p. 1081. doi: 10.1158/1055-9965.EPI-07-0377. [DOI] [PubMed] [Google Scholar]
- 41.Bajaj A, Driver JA, Schernhammer ES. Parkinson’s disease and cancer risk: a systematic review and meta-analysis. Cancer Causes and Control. 2010;21(5):697–707. doi: 10.1007/s10552-009-9497-6. [DOI] [PubMed] [Google Scholar]
- 42.Ferreira JJ, Neutel D, Mestre T, et al. Skin cancer and Parkinson’s disease. Movement Disorders. 2010;25(2):139–148. doi: 10.1002/mds.22855. [DOI] [PubMed] [Google Scholar]
- 43.Garber K. Parkinson’s disease and cancer: the unexplored connection. Journal of the National Cancer Institute. 2010;102(6):371–374. doi: 10.1093/jnci/djq081. [DOI] [PubMed] [Google Scholar]
- 44.Ren H, Fu K, Mu C, Li B, Wang D, Wang G. DJ-1, a cancer and Parkinson’s disease associated protein, regulates autophagy through JNK pathway in cancer cells. Cancer Letters. 2010;297(1):101–108. doi: 10.1016/j.canlet.2010.05.001. [DOI] [PubMed] [Google Scholar]
- 45.Crespi B. Autism and cancer risk. Autism Research. 2011;4:302–310. doi: 10.1002/aur.208. [DOI] [PubMed] [Google Scholar]
- 46.Sund R, Pukkala E, Patja K. Cancer incidence among persons with fragile X syndrome in Finland: a population-based study. Journal of Intellectual Disability Research. 2009;53(1):85–90. doi: 10.1111/j.1365-2788.2008.01116.x. [DOI] [PubMed] [Google Scholar]
- 47.Bartkova J, Zemanova M, Bartek J. Expression of CDK7/CAK in normal and tumour cells of diverse histogenesis, cell-cycle position and differentiation. International Journal of Cancer. 1996;66(6):732–737. doi: 10.1002/(SICI)1097-0215(19960611)66:6<732::AID-IJC4>3.0.CO;2-0. [DOI] [PubMed] [Google Scholar]
- 48.Ishikaw M, Ogihara Y, Miura M. Visualization of radiation-induced cell cycle-associated events in tumor cells expressing the fusion protein of Azami Green and the destruction box of human Geminin. Biochemical and Biophysical Research Communications. 2009;389(3):426–430. doi: 10.1016/j.bbrc.2009.08.160. [DOI] [PubMed] [Google Scholar]
- 49.Smith PJ, Marquez N, Wiltshire M, et al. Mitotic bypass via an occult cell cycle phase following DNA topoisomerase II inhibition in p53 functional human tumor cells. Cell Cycle. 2007;6(16):2071–2081. doi: 10.4161/cc.6.16.4585. [DOI] [PubMed] [Google Scholar]
- 50.Nagy Z, Esiri MM, Cato AM, Smith AD. Cell cycle markers in the hippocampus in Alzheimer’s disease. Acta Neuropathologica. 1997;94(1):6–15. doi: 10.1007/s004010050665. [DOI] [PubMed] [Google Scholar]
- 51.McShea A, Harris PLR, Webster KR, Wahl AF, Smith MA. Abnormal expression of the cell cycle regulators P16 and CDK4 in Alzheimer’s disease. American Journal of Pathology. 1997;150(6):1933–1939. [PMC free article] [PubMed] [Google Scholar]
- 52.Busser J, Geldmacher DS, Herrup K. Ectopic cell cycle proteins predict the sites of neuronal cell death in Alzheimer’s disease brain. Journal of Neuroscience. 1998;18(8):2801–2807. doi: 10.1523/JNEUROSCI.18-08-02801.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Yang Y, Geldmacher DS, Herrup K. DNA replication precedes neuronal cell death in Alzheimer’s disease. Journal of Neuroscience. 2001;21(8):2661–2668. doi: 10.1523/JNEUROSCI.21-08-02661.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Yang Y, Mufson EJ, Herrup K. Neuronal cell death is preceded by cell cycle events at all stages of Alzheimer’s disease. Journal of Neuroscience. 2003;23(7):2557–2563. doi: 10.1523/JNEUROSCI.23-07-02557.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Jordan-Sciutto KL, Dorsey R, Chalovich EM, Hammond RR, Achim CL. Expression patterns of retinoblastoma protein in Parkinson disease. Journal of Neuropathology and Experimental Neurology. 2003;62(1):68–74. doi: 10.1093/jnen/62.1.68. [DOI] [PubMed] [Google Scholar]
- 56.Nagy Z, Esiri MM. Neuronal cyclin expression in the hippocampus in temporal lobe epilepsy. Experimental Neurology. 1998;150(2):240–247. doi: 10.1006/exnr.1997.6753. [DOI] [PubMed] [Google Scholar]
- 57.Ranganathan S, Bowser R. Alterations in G1 to S phase cell-cycle regulators during amyotrophic lateral sclerosis. American Journal of Pathology. 2003;162(3):823–835. doi: 10.1016/S0002-9440(10)63879-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Herrup K, Neve R, Ackerman SL, Copani A. Divide and die: cell cycle events as triggers of nerve cell death. Journal of Neuroscience. 2004;24(42):9232–9239. doi: 10.1523/JNEUROSCI.3347-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Yang Y, Varvel NH, Lamb BT, Herrup K. Ectopic cell cycle events link human Alzheimer’s disease and amyloid precursor protein transgenic mouse models. Journal of Neuroscience. 2006;26(3):775–784. doi: 10.1523/JNEUROSCI.3707-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Khurana V, Lu Y, Steinhilb ML, Oldham S, Shulman JM, Feany MB. TOR-mediated cell-cycle activation causes neurodegeneration in a Drosophila tauopathy model. Current Biology. 2006;16(3):230–241. doi: 10.1016/j.cub.2005.12.042. [DOI] [PubMed] [Google Scholar]
- 61.Nguyen MD, Boudreau M, Kriz J, Couillard-Després S, Kaplan DR, Julien JP. Cell cycle regulators in the neuronal death pathway of amyotrophic lateral sclerosis caused by mutant superoxide dismutase 1. Journal of Neuroscience. 2003;23(6):2131–2140. doi: 10.1523/JNEUROSCI.23-06-02131.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Imai H, Harland J, McCulloch J, Graham DI, Brown SM, Macrae IM. Specific expression of the cell cycle regulation proteins, GADD34 and PCNA, in the peri-infarct zone after focal cerebral ischaemia in the rat. European Journal of Neuroscience. 2002;15(12):1929–1936. doi: 10.1046/j.1460-9568.2002.02025.x. [DOI] [PubMed] [Google Scholar]
- 63.O’Hare M, Wang F, Park DS. Cyclin-dependent kinases as potential targets to improve stroke outcome. Pharmacology and Therapeutics. 2002;93(2-3):135–143. doi: 10.1016/s0163-7258(02)00183-3. [DOI] [PubMed] [Google Scholar]
- 64.Kuan CY, Schloemer AJ, Lu A, et al. Hypoxia-ischemia induces DNA synthesis without cell proliferation in dying neurons in adult rodent brain. Journal of Neuroscience. 2004;24(47):10763–10772. doi: 10.1523/JNEUROSCI.3883-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Veeriah S, Taylor BS, Meng S, et al. Somatic mutations of the Parkinson’s disease-associated gene PARK2 in glioblastoma and other human malignancies. Nature Genetics. 2010;42(1):77–82. doi: 10.1038/ng.491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Park DS, Farinell SE, Greene LA. Inhibitors of cyclin-dependent kinases promote survival of post-mitotic neuronally differentiated PC12 cells and sympathetic neurons. Journal of Biological Chemistry. 1996;271(14):8161–8169. doi: 10.1074/jbc.271.14.8161. [DOI] [PubMed] [Google Scholar]
- 67.Park DS, Morris EJ, Greene LA, Geller HM. G1/S cell cycle blockers and inhibitors of cyclin-dependent kinases suppress camptothecin-induced neuronal apoptosis. Journal of Neuroscience. 1997;17(4):1256–1270. doi: 10.1523/JNEUROSCI.17-04-01256.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Copani A, Condorelli F, Caruso A, et al. Mitotic signaling by β-amyloid causes neuronal death. FASEB Journal. 1999;13:2225–2234. [PubMed] [Google Scholar]
- 69.Varvel NH, Bhaskar K, Patil AR, Pimplikar SW, Herrup K, Lamb BT. Aβ oligomers induce neuronal cell cycle events in Alzheimer’s disease. Journal of Neuroscience. 2008;28(43):10786–10793. doi: 10.1523/JNEUROSCI.2441-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Williamson R, Scales T, Clark BR, et al. Rapid tyrosine phosphorylation of neuronal proteins including tau and focal adhesion kinase in response to amyloid-β peptide exposure: involvement of Src family protein kinases. Journal of Neuroscience. 2002;22(1):10–20. doi: 10.1523/JNEUROSCI.22-01-00010.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Wang H, Reiser G. Thrombin signaling in the brain: the role of protease-activated receptors. Biological Chemistry. 2003;384(2):193–202. doi: 10.1515/BC.2003.021. [DOI] [PubMed] [Google Scholar]
- 72.Ohnishi M, Katsuki H, Fujimoto S, Takagi M, Kume T, Akaike A. Involvement of thrombin and mitogen-activated protein kinase pathways in hemorrhagic brain injury. Experimental Neurology. 2007;206(1):43–52. doi: 10.1016/j.expneurol.2007.03.030. [DOI] [PubMed] [Google Scholar]
- 73.Guan J, Luo Y, Denker BM. Purkinje cell protein-2 (Pcp2) stimulates differentiation in PC12 cells by Gβγ-mediated activation of Ras and p38 MAPK. Biochemical Journal. 2005;392(2):389–397. doi: 10.1042/BJ20042102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Segarra J, Balenci L, Drenth T, Maina F, Lamballe F. Combined signaling through ERK, PI3K/AKT, and RAC1/p38 is required for Met-triggered cortical neuron migration. Journal of Biological Chemistry. 2006;281(8):4771–4778. doi: 10.1074/jbc.M508298200. [DOI] [PubMed] [Google Scholar]
- 75.Lopez-Bergami P, Ronai Z. Requirements for PKC-augmented JNK activation by MKK4/7. International Journal of Biochemistry and Cell Biology. 2008;40(5):1055–1064. doi: 10.1016/j.biocel.2007.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Dwivedi Y, Pandey GN. Effects of treatment with haloperidol, chlorpromazine, and clozapine on protein kinase C (PKC) and phosphoinositide-specific phospholipase C (PI-PLC) activity and on mRNA and protein expression of PKC and PLC isozymes in rat brain. Journal of Pharmacology and Experimental Therapeutics. 1999;291(2):688–704. [PubMed] [Google Scholar]
- 77.Zhu X, Rottkamp CA, Boux H, Takeda A, Perry G, Smith MA. Activation of p38 kinase links tau phosphorylation, oxidative stress, and cell cycle-related events in Alzheimer disease. Journal of Neuropathology and Experimental Neurology. 2000;59(10):880–888. doi: 10.1093/jnen/59.10.880. [DOI] [PubMed] [Google Scholar]
- 78.Xing B, Xin T, Hunter RL, Bing G. Pioglitazone inhibition of lipopolysaccharide-induced nitric oxide synthase is associated with altered activity of p38 MAP kinase and PI3K/ Akt. Journal of Neuroinflammation. 2008;5, article 4 doi: 10.1186/1742-2094-5-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Tan J, et al. Neurodegenerative disease treatment using jAK/STAT inhibition. PCT/US2008/055646, 2008.
- 80.Goody RJ, Beckham JD, Rubtsova K, Tyler KL. JAK-STAT signaling pathways are activated in the brain following reovirus infection. Journal of NeuroVirology. 2007;13(4):373–383. doi: 10.1080/13550280701344983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Li N, Wang C, Wu Y, Liu X, Cao X. Ca2+/calmodulin-dependent protein kinase II promotes cell cycle progression by directly activating MEK1 and subsequently modulating p27 phosphorylation. Journal of Biological Chemistry. 2009;284(5):3021–3027. doi: 10.1074/jbc.M805483200. [DOI] [PubMed] [Google Scholar]
- 82.Mao LM, Tang QS, Wang JQ. Regulation of extracellular signal-regulated kinase phosphorylation in cultured rat striatal neurons. Brain Research Bulletin. 2009;78(6):328–334. doi: 10.1016/j.brainresbull.2008.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.De Bernardo S, Canals S, Casarejos MJ, Solano RM, Menendez J, Mena MA. Role of extracellular signal-regulated protein kinase in neuronal cell death induced by glutathione depletion in neuron/glia mesencephalic cultures. Journal of Neurochemistry. 2004;91(3):667–682. doi: 10.1111/j.1471-4159.2004.02744.x. [DOI] [PubMed] [Google Scholar]
- 84.Copani A, Nicoletti F. Cell-Cycle Mechanisms and Neuronal Cell Death. New York, NY, USA: Kluwer Academic Publishers/Plenum; 2005. [Google Scholar]
- 85.Qian JY, Leung A, Harding P, LaPointe MC. PGE2 stimulates human brain natriuretic peptide expression via EP4 and p42/44 MAPK. American Journal of Physiology. 2006;290(5):H1740–H1746. doi: 10.1152/ajpheart.00904.2005. [DOI] [PubMed] [Google Scholar]
- 86.Fiebich BL, Schleicher S, Spleiss O, Czygan M, Hüll M. Mechanisms of prostaglandin E2-induced interleukin-6 release in astrocytes: possible involvement of EP4-like receptors, p38 mitogen-activated protein kinase and protein kinase C. Journal of Neurochemistry. 2001;79(5):950–958. doi: 10.1046/j.1471-4159.2001.00652.x. [DOI] [PubMed] [Google Scholar]
- 87.Darmoul D, Gratio V, Devaud H, Lehy T, Laburthe M. Aberrant expression and activation of the thrombin receptor protease-activated receptor-1 induces cell proliferation and motility in human colon cancer cells. American Journal of Pathology. 2003;162(5):1503–1513. doi: 10.1016/S0002-9440(10)64283-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Traby L, Kaider A, Schmid R, et al. The effects of low-molecular-weight heparin at two different dosages on thrombin generation in cancer patients: a randomised controlled trial. Thrombosis and Haemostasis. 2010;104(1):92–99. doi: 10.1160/TH09-12-0863. [DOI] [PubMed] [Google Scholar]
- 89.Snyder KM, Kessler CM. The pivotal role of thrombin in cancer biology and tumorigenesis. Seminars in Thrombosis and Hemostasis. 2008;34(8):734–741. [Google Scholar]
- 90.Wang H, Reiser G. The role of the Ca2+-sensitive tyrosine kinase Pyk2 and Src in thrombin signalling in rat astrocytes. Journal of Neurochemistry. 2003;84(6):1349–1357. doi: 10.1046/j.1471-4159.2003.01637.x. [DOI] [PubMed] [Google Scholar]
- 91.Liu DZ, Cheng XY, Ander BP, et al. Src kinase inhibition decreases thrombin-induced injury and cell cycle re-entry in striatal neurons. Neurobiology of Disease. 2008;30(2):201–211. doi: 10.1016/j.nbd.2008.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Fujimoto S, Katsuki H, Ohnishi M, Takagi M, Kume T, Akaike A. Thrombin induces striatal neurotoxicity depending on mitogen-activated protein kinase pathways in vivo. Neuroscience. 2007;144(2):694–701. doi: 10.1016/j.neuroscience.2006.09.049. [DOI] [PubMed] [Google Scholar]
- 93.Driessens G, Harsan L, Browaeys P, Giannakopoulos X, Velu T, Bruyns C. Assessment of in vivo chemotherapy-induced DNa damage in a p53-mutated rat tumor by micronuclei assay. Annals of the New York Academy of Sciences. 2003;1010:775–779. doi: 10.1196/annals.1299.139. [DOI] [PubMed] [Google Scholar]
- 94.Van Kaick G, Delorme S. Therapy-induced effects in normal tissue. Radiologe. 2008;48(9):871–880. doi: 10.1007/s00117-008-1729-3. [DOI] [PubMed] [Google Scholar]
- 95.Lucas K, Gula MJ, Blatt J. Relapse in acute lymphoblastic leukemia as a function of white blood cell and absolute neutrophil counts during maintenance chemotherapy. Pediatric Hematology and Oncology. 1992;9(2):91–97. doi: 10.3109/08880019209018324. [DOI] [PubMed] [Google Scholar]
- 96.Wefel JS, Saleeba AK, Buzdar AU, Meyers CA. Acute and late onset cognitive dysfunction associated with chemotherapy in women with breast cancer. Cancer. 2010;116(14):3348–3356. doi: 10.1002/cncr.25098. [DOI] [PubMed] [Google Scholar]
- 97.Harrington CB, Hansen JA, Moskowitz M, Todd BL, Feuerstein M. It’s not over when it’s over: long-term symptoms in cancer survivors—a systematic review. International Journal of Psychiatry in Medicine. 2010;40(2):163–181. doi: 10.2190/PM.40.2.c. [DOI] [PubMed] [Google Scholar]
- 98.Dietrich J, Monje M, Wefel J, Meyers C. Clinical patterns and biological correlates of cognitive dysfunction associated with cancer therapy. Oncologist. 2008;13(12):1285–1295. doi: 10.1634/theoncologist.2008-0130. [DOI] [PubMed] [Google Scholar]
- 99.Taphoorn MJB, Klein M. Cognitive deficits in adult patients with brain tumours. Lancet Neurology. 2004;3(3):159–168. doi: 10.1016/S1474-4422(04)00680-5. [DOI] [PubMed] [Google Scholar]
- 100.Byrne TN. Cognitive sequelae of brain tumor treatment. Current Opinion in Neurology. 2005;18(6):662–666. doi: 10.1097/01.wco.0000191511.52745.99. [DOI] [PubMed] [Google Scholar]
- 101.Brière M-E, Scott JG, McNall-Knapp RY, Adams RL. Cognitive outcome in pediatric brain tumor survivors: delayed attention deficit at long-term follow-up. Pediatric Blood and Cancer. 2008;50(2):337–340. doi: 10.1002/pbc.21223. [DOI] [PubMed] [Google Scholar]
- 102.Hilverda K, Bosma I, Heimans JJ, et al. Cognitive functioning in glioblastoma patients during radiotherapy and temozolomide treatment: initial findings. Journal of Neuro-Oncology. 2010;97(1):89–94. doi: 10.1007/s11060-009-9993-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Talacchi A, Santini B, Savazzi S, Gerosa M. Cognitive effects of tumour and surgical treatment in glioma patients. Journal of Neuro-Oncology. 2010;103(3):541–549. doi: 10.1007/s11060-010-0417-0. [DOI] [PubMed] [Google Scholar]
- 104.Correa DD. Cognitive functions in brain tumor patients. Hematology/Oncology Clinics of North America. 2006;20(6):1363–1376. doi: 10.1016/j.hoc.2006.09.012. [DOI] [PubMed] [Google Scholar]
- 105.Marsh JC, Gielda BT, Herskovic AM, Abrams RA. Cognitive sparing during the administration of whole brain radiotherapy and prophylactic cranial irradiation: current concepts and approaches. Journal of Oncology. 2010;2010:16 pages. doi: 10.1155/2010/198208. Article ID 198208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Brandt MD, Storch A. Neurogenesis in the adult brain: from bench to bedside? Fortschritte der Neurologie Psychiatrie. 2008;76(9):517–529. doi: 10.1055/s-2008-1038218. [DOI] [PubMed] [Google Scholar]
- 107.Shen J, Xie L, Mao X, et al. Neurogenesis after primary intracerebral hemorrhage in adult human brain. Journal of Cerebral Blood Flow and Metabolism. 2008;28(8):1460–1468. doi: 10.1038/jcbfm.2008.37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Neundörfer B. Does the neurogenesis in the adult brain show the way into the future? Fortschritte der Neurologie Psychiatrie. 2008;76(9):p. 511. doi: 10.1055/s-2008-1038252. [DOI] [PubMed] [Google Scholar]
- 109.Liu J, Solway K, Messing RO, Sharp FR. Increased neurogenesis in the dentate gyrus after transient global ischemia in gerbils. Journal of Neuroscience. 1998;18(19):7768–7778. doi: 10.1523/JNEUROSCI.18-19-07768.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Abdipranoto A, Wu S, Stayte S, Vissel B. The role of neurogenesis in neurodegenerative diseases and its implications for therapeutic development. CNS and Neurological Disorders. 2008;7(2):187–210. doi: 10.2174/187152708784083858. [DOI] [PubMed] [Google Scholar]
- 111.Chesnokova V, Pechnick RN. Antidepressants and Cdk inhibitors: releasing the brake on neurogenesis? Cell Cycle. 2008;7(15):2321–2326. doi: 10.4161/cc.6446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Ohab JJ, Fleming S, Blesch A, Carmichael ST. A neurovascular niche for neurogenesis after stroke. Journal of Neuroscience. 2006;26(50):13007–13016. doi: 10.1523/JNEUROSCI.4323-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Palmer TD, Willhoite AR, Gage FH. Vascular niche for adult hippocampal neurogenesis. Journal of Comparative Neurology. 2000;425(4):479–494. doi: 10.1002/1096-9861(20001002)425:4<479::aid-cne2>3.0.co;2-3. [DOI] [PubMed] [Google Scholar]
- 114.Eriksson PS, Perfilieva E, Björk-Eriksson T, et al. Neurogenesis in the adult human hippocampus. Nature Medicine. 1998;4(11):1313–1317. doi: 10.1038/3305. [DOI] [PubMed] [Google Scholar]
- 115.Roy NS, Wang S, Jiang L, et al. In vitro neurogenesis by progenitor cells isolated from the adult human hippocampus. Nature Medicine. 2000;6(3):271–277. doi: 10.1038/73119. [DOI] [PubMed] [Google Scholar]
- 116.Doetsch F, Caille I, Lim DA, Garcia-Verdugo JM, Alvarez-Buylla A. Subventricular zone astrocytes are neural stem cells in the adult mammalian brain. Cell. 1999;97(6):703–716. doi: 10.1016/s0092-8674(00)80783-7. [DOI] [PubMed] [Google Scholar]
- 117.Morshead CM, Reynolds BA, Craig CG, et al. Neural stem cells in the adult mammalian forebrain: a relatively quiescent subpopulation of subependymal cells. Neuron. 1994;13(5):1071–1082. doi: 10.1016/0896-6273(94)90046-9. [DOI] [PubMed] [Google Scholar]
- 118.Liu D-Z, Ander BP, Xu H, et al. Blood-brain barrier breakdown and repair by Src after thrombin-induced injury. Annals of Neurology. 2010;67(4):526–533. doi: 10.1002/ana.21924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Becker S. A computational principle for hippocampal learning and neurogenesis. Hippocampus. 2005;15(6):722–738. doi: 10.1002/hipo.20095. [DOI] [PubMed] [Google Scholar]
- 120.Aimone JB, Wiles J, Gage FH. Potential role for adult neurogenesis in the encoding of time in new memories. Nature Neuroscience. 2006;9(6):723–727. doi: 10.1038/nn1707. [DOI] [PubMed] [Google Scholar]
- 121.Fallon J, Reid S, Kinyamu R, et al. In vivo induction of massive proliferation, directed migration, and differentiation of neural cells in the adult mammalian brain. Proceedings of the National Academy of Sciences of the United States of America. 2000;97(26):14686–14691. doi: 10.1073/pnas.97.26.14686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Schneider LS, Dagerman KS, Higgins JPT, McShane R. Lack of evidence for the efficacy of memantine in mild Alzheimer disease. Archives of Neurology. 2011;68(8):991–998. doi: 10.1001/archneurol.2011.69. [DOI] [PubMed] [Google Scholar]
- 123.Suzuki M, Nelson AD, Eickstaedt JB, Wallace K, Wright LS, Svendsen CN. Glutamate enhances proliferation and neurogenesis in human neural progenitor cell cultures derived from the fetal cortex. European Journal of Neuroscience. 2006;24(3):645–653. doi: 10.1111/j.1460-9568.2006.04957.x. [DOI] [PubMed] [Google Scholar]
- 124.Zhang X, Yeung ED, Wang J, et al. Isoliquiritigenin, a natural anti-oxidant, selectively inhibits the proliferation of prostate cancer cells. Clinical and Experimental Pharmacology and Physiology. 2010;37(8):841–847. doi: 10.1111/j.1440-1681.2010.05395.x. [DOI] [PubMed] [Google Scholar]
- 125.Abe K. Neuroprotective therapy for ischemic stroke with free radical scavenger and gene-stem cell therapy. Clinical Neurology. 2008;48(11):896–898. doi: 10.5692/clinicalneurol.48.896. [DOI] [PubMed] [Google Scholar]
- 126.Wang CX, Shuaib A. Neuroprotective effects of free radical scavengers in stroke. Drugs and Aging. 2007;24(7):537–546. doi: 10.2165/00002512-200724070-00002. [DOI] [PubMed] [Google Scholar]
- 127.Eerman K, Brodaty H. Tocopherol (vitamin E) in Alzheimer’s disease and other neurodegenerative disorders. CNS Drugs. 2004;18(12):807–825. doi: 10.2165/00023210-200418120-00005. [DOI] [PubMed] [Google Scholar]
- 128.Viña J, Lloret A, Ortí R, Alonso D. Molecular bases of the treatment of Alzheimer’s disease with antioxidants: prevention of oxidative stress. Molecular Aspects of Medicine. 2004;25(1-2):117–123. doi: 10.1016/j.mam.2004.02.013. [DOI] [PubMed] [Google Scholar]
- 129.Park KW, Jin BK. Thrombin-induced oxidative stress contributes to the death of hippocampal neurons: role of neuronal NADPH oxidase. Journal of Neuroscience Research. 2008;86(5):1053–1063. doi: 10.1002/jnr.21571. [DOI] [PubMed] [Google Scholar]
- 130.Levy G, Kaufmann P, Buchsbaum R, et al. A two-stage design for a phase II clinical trial of coenzyme Q10 in ALS. Neurology. 2006;66(5):660–663. doi: 10.1212/01.wnl.0000201182.60750.66. [DOI] [PubMed] [Google Scholar]
- 131.Ferrante KL, Shefner J, Zhang H, et al. Tolerance of high-dose (3,000 mg/day) coenzyme Q10 in ALS. Neurology. 2005;65(11):1834–1836. doi: 10.1212/01.wnl.0000187070.35365.d7. [DOI] [PubMed] [Google Scholar]
- 132.Yoshino H, Kimura A. Investigation of the therapeutic effects of edaravone, a free radical scavenger, on amyotrophic lateral sclerosis (Phase II study). Amyotrophic Lateral Sclerosis. 2006;7(4):241–245. doi: 10.1080/17482960600881870. [DOI] [PubMed] [Google Scholar]
- 133.Ito H, Wate R, Zhang J, et al. Treatment with edaravone, initiated at symptom onset, slows motor decline and decreases SOD1 deposition in ALS mice. Experimental Neurology. 2008;213(2):448–455. doi: 10.1016/j.expneurol.2008.07.017. [DOI] [PubMed] [Google Scholar]
- 134.Yuan WJ, Yasuhara T, Shingo T, et al. Neuroprotective effects of edaravone-administration on 6-OHDA-treated dopaminergic neurons. BMC Neuroscience. 2008;9, article 75 doi: 10.1186/1471-2202-9-75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Tanabe M, Nagatani Y, Saitoh K, Takasu K, Ono H. Pharmacological assessments of nitric oxide synthase isoforms and downstream diversity of NO signaling in the maintenance of thermal and mechanical hypersensitivity after peripheral nerve injury in mice. Neuropharmacology. 2009;56(3):702–708. doi: 10.1016/j.neuropharm.2008.12.003. [DOI] [PubMed] [Google Scholar]
- 136.Vanella L, Di Giacomo C, Acquaviva R, et al. The DDAH/NOS pathway in human prostatic cancer cell lines: antiangiogenic effect of L-NAME. International Journal of Oncology. 2011;39(5):1303–1310. doi: 10.3892/ijo.2011.1107. [DOI] [PubMed] [Google Scholar]
- 137.Lee DY, Park KW, Jin BK. Thrombin induces neurodegeneration and microglial activation in the cortex in vivo and in vitro: proteolytic and non-proteolytic actions. Biochemical and Biophysical Research Communications. 2006;346(3):727–738. doi: 10.1016/j.bbrc.2006.05.174. [DOI] [PubMed] [Google Scholar]
- 138.Sabichi AL, Lee JJ, Grossman HB, et al. A randomized controlled trial of celecoxib to prevent recurrence of non-muscle-invasive bladder cancer. Cancer Prevention Research. 2011;4(10):1580–1589. doi: 10.1158/1940-6207.CAPR-11-0036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Koch A, Bergman B, Holmberg E, et al. Effect of celecoxib on survival in patients with advanced non-small cell lung cancer: a double blind randomised clinical phase III trial (CYCLUS study) by the Swedish Lung Cancer Study Group. European Journal of Cancer. 2011;47(10):1546–1555. doi: 10.1016/j.ejca.2011.03.035. [DOI] [PubMed] [Google Scholar]
- 140.Gridelli C, Gallo C, Ceribelli A, et al. Factorial phase III randomised trial of rofecoxib and prolonged constant infusion of gemcitabine in advanced non-small-cell lung cancer: the GEmcitabine-COxib in NSCLC (GECO) study. Lancet Oncology. 2007;8(6):500–512. doi: 10.1016/S1470-2045(07)70146-8. [DOI] [PubMed] [Google Scholar]
- 141.Kao J, Genden EM, Chen C-T, et al. Phase 1 trial of concurrent erlotinib, celecoxib, and reirradiation for recurrent head and neck cancer. Cancer. 2011;117(14):3173–3181. doi: 10.1002/cncr.25786. [DOI] [PubMed] [Google Scholar]
- 142.Lipton A, Campbell-Baird C, Witters L, Harvey H, Ali S. Phase II trial of gemcitabine, irinotecan, and celecoxib in patients with advanced pancreatic cancer. Journal of Clinical Gastroenterology. 2010;44(4):286–288. doi: 10.1097/MCG.0b013e3181cda097. [DOI] [PubMed] [Google Scholar]
- 143.Antonarakis ES, Heath EI, Walczak JR, et al. Phase II, randomized, placebo-controlled trial of neoadjuvant celecoxib in men with clinically localized prostate cancer: evaluation of drug-specific biomarkers. Journal of Clinical Oncology. 2009;27(30):4986–4993. doi: 10.1200/JCO.2009.21.9410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Fabi A, Metro G, Papaldo P, et al. Impact of celecoxib on capecitabine tolerability and activity in pretreated metastatic breast cancer: results of a phase II study with biomarker evaluation. Cancer Chemotherapy and Pharmacology. 2008;62(4):717–725. doi: 10.1007/s00280-007-0650-1. [DOI] [PubMed] [Google Scholar]
- 145.Midgley RS, McConkey CC, Johnstone EC, et al. Phase III randomized trial assessing rofecoxib in the adjuvant setting of colorectal cancer: final results of the VICTOR trial. Journal of Clinical Oncology. 2010;28(30):4575–4580. doi: 10.1200/JCO.2010.29.6244. [DOI] [PubMed] [Google Scholar]
- 146.Youns M, Efferth T, Hoheisel JD. Transcript profiling identifies novel key players mediating the growth inhibitory effect of NS-398 on human pancreatic cancer cells. European Journal of Pharmacology. 2011;650(1):170–177. doi: 10.1016/j.ejphar.2010.10.026. [DOI] [PubMed] [Google Scholar]
- 147.Fernández-Martínez AB, Bajo AM, Valdehita A, et al. Multifunctional role of VIP in prostate cancer progression in a xenograft model: suppression by curcumin and COX-2 inhibitor NS-398. Peptides. 2009;30(12):2357–2364. doi: 10.1016/j.peptides.2009.09.018. [DOI] [PubMed] [Google Scholar]
- 148.Zhang L, Tu J, Yu Z-L, Wu Y-D, Xu C-M, Zhang S-T. Effects of the inhibition of cyclooxygenase-2 on human esophageal cancer cells: inhibition of cell proliferation and induction of apoptosis. Pathology and Oncology Research. 2010;16(1):39–45. doi: 10.1007/s12253-009-9185-6. [DOI] [PubMed] [Google Scholar]
- 149.Banu N, Buda A, Chell S, et al. Inhibition of COX-2 with NS-398 decreases colon cancer cell motility through blocking epidermal growth factor receptor transactivation: possibilities for combination therapy. Cell Proliferation. 2007;40(5):768–779. doi: 10.1111/j.1365-2184.2007.00459.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Warner TD, Mitchell JA. Cyclooxygenases: new forms, new inhibitors, and lessons from the clinic. FASEB Journal. 2004;18(7):790–804. doi: 10.1096/fj.03-0645rev. [DOI] [PubMed] [Google Scholar]
- 151.Etminan M, Gill S, Samii A. Effect of non-steroidal anti-inflammatory drugs on risk of Alzheimer’s disease: systematic review and meta-analysis of observational studies. British Medical Journal. 2003;327(7407):128–131. doi: 10.1136/bmj.327.7407.128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Heo JH, Seo HN, Choe YJ, et al. T-type Ca2+ channel blockers suppress the growth of human cancer cells. Bioorganic and Medicinal Chemistry Letters. 2008;18(14):3899–3901. doi: 10.1016/j.bmcl.2008.06.034. [DOI] [PubMed] [Google Scholar]
- 153.Franke CL, Palm R, Dalby M, et al. Flunarizine in stroke treatment (FIST): a double-blind, placebo-controlled trial in Scandinavia and the Netherlands. Acta Neurologica Scandinavica. 1996;93(1):56–60. doi: 10.1111/j.1600-0404.1996.tb00171.x. [DOI] [PubMed] [Google Scholar]
- 154.North WG, Gao G, Memoli VA, Pang RH, Lynch L. Breast cancer expresses functional NMDA receptors. Breast Cancer Research and Treatment. 2010;122(2):307–314. doi: 10.1007/s10549-009-0556-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Yang H, Chopp M, Jiang F, Zhang X, Schallert T. Interruption of functional recovery by the NMDA glutamate antagonist MK801 after compression of the sensorimotor cortex: implications for treatment of tumors or other mass-related brain injuries. Experimental Neurology. 2006;200(1):262–266. doi: 10.1016/j.expneurol.2006.02.007. [DOI] [PubMed] [Google Scholar]
- 156.Andrews J. Amyotrophic lateral sclerosis: clinical management and research update. Current Neurology and Neuroscience Reports. 2009;9(1):59–68. doi: 10.1007/s11910-009-0010-0. [DOI] [PubMed] [Google Scholar]
- 157.McIntosh TK, Smith DH, Voddi M, Perri BR, Stutzmann J-M. Riluzole, a novel neuroprotective agent, attenuates both neurologic motor and cognitive dysfunction following experimental brain injury in the rat. Journal of Neurotrauma. 1996;13(12):767–780. doi: 10.1089/neu.1996.13.767. [DOI] [PubMed] [Google Scholar]
- 158.Belayev L, Alonso OF, Liu Y, et al. Talampanel, a novel noncompetitive AMPA antagonist, is neuroprotective after traumatic brain injury in rats. Journal of Neurotrauma. 2001;18(10):1031–1038. doi: 10.1089/08977150152693728. [DOI] [PubMed] [Google Scholar]
- 159.Kitzman PH. Effectiveness of riluzole in suppressing spasticity in the spinal cord injured rat. Neuroscience Letters. 2009;455(2):150–153. doi: 10.1016/j.neulet.2009.03.016. [DOI] [PubMed] [Google Scholar]
- 160.Ardizzone TD, Lu A, Wagner KR, Tang Y, Ran R, Sharp FR. Glutamate receptor blockade attenuates glucose hypermetabolism in perihematomal brain after experimental intracerebral hemorrhage in rat. Stroke. 2004;35(11):2587–2591. doi: 10.1161/01.STR.0000143451.14228.ff. [DOI] [PubMed] [Google Scholar]
- 161.Meden P, Overgaard K, Sereghy T, Boysen G. Enhancing the efficacy of thrombolysis by AMPA receptor blockade with NBQX in a rat embolic stroke model. Journal of the Neurological Sciences. 1993;119(2):209–216. doi: 10.1016/0022-510x(93)90136-m. [DOI] [PubMed] [Google Scholar]
- 162.Watanabe K, Kanno T, Oshima T, Miwa H, Tashiro C, Nishizaki T. The NMDA receptor NR2A subunit regulates proliferation of MKN45 human gastric cancer cells. Biochemical and Biophysical Research Communications. 2008;367(2):487–490. doi: 10.1016/j.bbrc.2007.12.167. [DOI] [PubMed] [Google Scholar]
- 163.Danysz W, Parsons CG. The NMDA receptor antagonist memantine as a symptomatological and neuroprotective treatment for Alzheimer’s disease: preclinical evidence. International Journal of Geriatric Psychiatry. 2003;18(1):S23–S32. doi: 10.1002/gps.938. [DOI] [PubMed] [Google Scholar]
- 164.Molinuevo JL, Lladó A, Rami L. Memantine: targeting glutamate excitotoxicity in Alzheimer's disease and other dementias. American Journal of Alzheimer's Disease and other Dementias. 2005;20(2):77–85. doi: 10.1177/153331750502000206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Robinson DM, Keating GM. Memantine: a review of its use in Alzheimer’s disease. Drugs. 2006;66(11):1515–1534. doi: 10.2165/00003495-200666110-00015. [DOI] [PubMed] [Google Scholar]
- 166.Iraqi A, Hughes TL. Nightmares and memantine: a case report and review of literature. Journal of the American Medical Directors Association. 2009;10(1):77–78. doi: 10.1016/j.jamda.2008.10.001. [DOI] [PubMed] [Google Scholar]
- 167.Zdanys K, Tampi RR. A systematic review of off-label uses of memantine for psychiatric disorders. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2008;32(6):1362–1374. doi: 10.1016/j.pnpbp.2008.01.008. [DOI] [PubMed] [Google Scholar]
- 168.Raghavendra Rao VL, Dogan A, Todd KG, Bowen KK, Dempsey RJ. Neuroprotection by memantine, a non-competitive NMDA receptor antagonist after traumatic brain injury in rats. Brain Research. 2001;911(1):96–100. doi: 10.1016/s0006-8993(01)02617-8. [DOI] [PubMed] [Google Scholar]
- 169.Lee S-T, Chu K, Jung K-H, et al. Memantine reduces hematoma expansion in experimental intracerebral hemorrhage, resulting in functional improvement. Journal of Cerebral Blood Flow and Metabolism. 2006;26(4):536–544. doi: 10.1038/sj.jcbfm.9600213. [DOI] [PubMed] [Google Scholar]
- 170.Babu CS, Ramanathan M. Pre-ischemic treatment with memantine reversed the neurochemical and behavioural parameters but not energy metabolites in middle cerebral artery occluded rats. Pharmacology Biochemistry and Behavior. 2009;92(3):424–432. doi: 10.1016/j.pbb.2009.01.010. [DOI] [PubMed] [Google Scholar]
- 171.Yu L, Garg HG, Li B, Linhardt RJ, Hales CA. Antitumor effect of butanoylated heparin with low anticoagulant activity on lung cancer growth in mice and rats. Current Cancer Drug Targets. 2010;10(2):229–241. doi: 10.2174/156800910791054176. [DOI] [PubMed] [Google Scholar]
- 172.Mikulík R, Dufek M, Goldemund D, Reif M. A pilot study on systemic thrombolysis followed by low molecular weight heparin in ischemic stroke. European Journal of Neurology. 2006;13(10):1106–1111. doi: 10.1111/j.1468-1331.2006.01458.x. [DOI] [PubMed] [Google Scholar]
- 173.Sun Z, Zhao Z, Zhao S, et al. Recombinant hirudin treatment modulates aquaporin-4 and aquaporin-9 expression after intracerebral hemorrhage in vivo. Molecular Biology Reports. 2009;36(5):1119–1127. doi: 10.1007/s11033-008-9287-3. [DOI] [PubMed] [Google Scholar]
- 174.Xue M, Hollenberg MD, Yong VW. Combination of thrombin and matrix metalloproteinase-9 exacerbates neurotoxicity in cell culture and intracerebral hemorrhage in mice. Journal of Neuroscience. 2006;26(40):10281–10291. doi: 10.1523/JNEUROSCI.2806-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Goerge T, Barg A, Schnaeker EM, et al. Tumor-derived matrix metalloproteinase-1 targets endothelial proteinase-activated receptor 1 promoting endothelial cell activation. Cancer Research. 2006;66(15):7766–7774. doi: 10.1158/0008-5472.CAN-05-3897. [DOI] [PubMed] [Google Scholar]
- 176.Junge CE, Sugawara T, Mannaioni G, et al. The contribution of protease-activated receptor 1 to neuronal damage caused by transient focal cerebral ischemia. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(22):13019–13024. doi: 10.1073/pnas.2235594100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Hamill CE, Caudle WM, Richardson JR, et al. Exacerbation of dopaminergic terminal damage in a mouse model of Parkinson’s disease by the G-protein-coupled receptor protease-activated receptor 1. Molecular Pharmacology. 2007;72(3):653–664. doi: 10.1124/mol.107.038158. [DOI] [PubMed] [Google Scholar]
- 178.Acosta MT, Kardel PG, Walsh KS, Rosenbaum KN, Gioia GA, Packer RJ. Lovastatin as treatment for neurocognitive deficits in neurofibromatosis type 1: phase I study. Pediatric Neurology. 2011;45(4):241–245. doi: 10.1016/j.pediatrneurol.2011.06.016. [DOI] [PubMed] [Google Scholar]
- 179.Knox JJ, Siu LL, Chen E, et al. A Phase I trial of prolonged administration of lovastatin in patients with recurrent or metastatic squamous cell carcinoma of the head and neck or of the cervix. European Journal of Cancer. 2005;41(4):523–530. doi: 10.1016/j.ejca.2004.12.013. [DOI] [PubMed] [Google Scholar]
- 180.Barkan B, Starinsky S, Friedman E, Stein R, Kloog Y. The Ras inhibitor farnesylthiosalicylic acid as a potential therapy for neurofibromatosis type 1. Clinical Cancer Research. 2006;12(18):5533–5542. doi: 10.1158/1078-0432.CCR-06-0792. [DOI] [PubMed] [Google Scholar]
- 181.Björkhem-Bergman L, Acimovic J, Torndal UB, Parini P, Eriksson LC. Lovastatin prevents carcinogenesis in a rat model for liver cancer. Effects of ubiquinone supplementation. Anticancer Research. 2010;30(4):1105–1112. [PubMed] [Google Scholar]
- 182.Martirosyan A, Clendening JW, Goard CA, Penn LZ. Lovastatin induces apoptosis of ovarian cancer cells and synergizes with doxorubicin: potential therapeutic relevance. BMC Cancer. 2010;10, article 103 doi: 10.1186/1471-2407-10-103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Klawitter J, Shokati T, Moll V, Christians U, Klawitter J. Effects of lovastatin on breast cancer cells: a proteo-metabonomic study. Breast Cancer Research. 2010;12(2, article R16) doi: 10.1186/bcr2485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Lee J, Lee I, Park C, Kang WK. Lovastatin-induced RhoA modulation and its effect on senescence in prostate cancer cells. Biochemical and Biophysical Research Communications. 2006;339(3):748–754. doi: 10.1016/j.bbrc.2005.11.075. [DOI] [PubMed] [Google Scholar]
- 185.Park IH, Kim JY, Jung JI, Han JY. Lovastatin overcomes gefitinib resistance in human non-small cell lung cancer cells with K-Ras mutations. Investigational New Drugs. 2010;28(6):791–799. doi: 10.1007/s10637-009-9319-4. [DOI] [PubMed] [Google Scholar]
- 186.Elkind MSV, Sacco RL, Macarthur RB, et al. The Neuroprotection with Statin Therapy for Acute Recovery Trial (NeuSTART): an adaptive design phase I dose-escalation study of high-dose lovastatin in acute ischemic stroke. International Journal of Stroke. 2008;3(3):210–218. doi: 10.1111/j.1747-4949.2008.00200.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Elkind MSV, Sacco RL, MacArthur RB, et al. High-dose lovastatin for acute ischemic stroke: results of the phase I dose escalation Neuroprotection with Statin Therapy for Acute Recovery Trial (NeuSTART) Cerebrovascular Diseases. 2009;28(3):266–275. doi: 10.1159/000228709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Smirnova IV, Citron BA, Arnold PM, Festoff BW. Neuroprotective signal transduction in model motor neurons exposed to thrombin: G-protein modulation effects on neurite outgrowth, Ca2+ mobilization, and apoptosis. Journal of Neurobiology. 2001;48(2):87–100. [PubMed] [Google Scholar]
- 189.Cunningham DD. Thrombin induces apoptosis in cultured neurons and astrocytes via a pathway requiring tyrosine kinase and RhoA activities. Journal of Neuroscience. 1997;17(14):5316–5326. doi: 10.1523/JNEUROSCI.17-14-05316.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Herold CI, Chadaram V, Peterson BL, et al. Phase II trial of dasatinib in patients with metastatic breast cancer using real-time pharmacodynamic tissue biomarkers of Src inhibition to escalate dosing. Clinical Cancer Research. 2011;17(18):6061–6070. doi: 10.1158/1078-0432.CCR-11-1071. [DOI] [PubMed] [Google Scholar]
- 191.Gucalp A, Sparano JA, Caravelli J, et al. Phase II trial of saracatinib (AZD0530), an oral SRC-inhibitor for the treatment of patients with hormone receptor-negative metastatic breast cancer. Clinical Breast Cancer. 2011;11(5):306–311. doi: 10.1016/j.clbc.2011.03.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Anbalagan M, Carrier L, Glodowski S, Hangauer D, Shan B, Rowan BG. KX-01, a novel Src kinase inhibitor directed toward the peptide substrate site, synergizes with tamoxifen in estrogen receptor alpha positive breast cancer. doi: 10.1007/s10549-011-1513-3. Breast Cancer Research and Treatment. In press. [DOI] [PubMed] [Google Scholar]
- 193.Pohorelic B, Singh R, Parkin S, et al. Role of Src in breast cancer cell migration and invasion in a breast cell/bone-derived cell microenvironment. doi: 10.1007/s10549-011-1753-2. Breast Cancer Research and Treatment. In press. [DOI] [PubMed] [Google Scholar]
- 194.Ceppi P, Rapa I, Lo Iacono M, et al. Expression and pharmacological inhibition of thymidylate synthase and Src kinase in nonsmall cell lung cancer. doi: 10.1002/ijc.26188. International Journal of Cancer. In press. [DOI] [PubMed] [Google Scholar]
- 195.Krystal GW, DeBerry CS, Linnekin D, Litz J. Lck associates with and is activated by kit in a small cell lung cancer cell line: inhibition of SCF-mediated growth by the Src family kinase inhibitor PP1. Cancer Research. 1998;58(20):4660–4666. [PubMed] [Google Scholar]
- 196.Kong L, Deng Z, Shen H, Zhang Y. Src family kinase inhibitor PP2 efficiently inhibits cervical cancer cell proliferation through down-regulating phospho-Src-Y416 and phospho-EGFR-Y1173. Molecular and Cellular Biochemistry. 2011;348(1-2):11–19. doi: 10.1007/s11010-010-0632-1. [DOI] [PubMed] [Google Scholar]
- 197.Fujimoto E, Sato H, Nagashima Y, et al. A Src family inhibitor (PP1) potentiates tumor-suppressive effect of connexin 32 gene in renal cancer cells. Life Sciences. 2005;76(23):2711–2720. doi: 10.1016/j.lfs.2004.10.049. [DOI] [PubMed] [Google Scholar]
- 198.Li L, Ren CH, Tahir SA, Ren C, Thompson TC. Caveolin-1 maintains activated Akt in prostate cancer cells through scaffolding domain binding site interactions with and inhibition of serine/threonine protein phosphatases PP1 and PP2A. Molecular and Cellular Biology. 2003;23(24):9389–9404. doi: 10.1128/MCB.23.24.9389-9404.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Ardizzone TD, Zhan X, Ander BP, Sharp FR. Src kinase inhibition improves acute outcomes after experimental intracerebral hemorrhage. Stroke. 2007;38(5):1621–1625. doi: 10.1161/STROKEAHA.106.478966. [DOI] [PubMed] [Google Scholar]
- 200.Siddiqui IA, Asim M, Hafeez BB, Adhami VM, Tarapore RS, Mukhtar H. Green tea polyphenol EGCG blunts androgen receptor function in prostate cancer. FASEB Journal. 2011;25(4):1198–1207. doi: 10.1096/fj.10-167924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Sen T, Dutta A, Chatterjee A. Epigallocatechin-3-gallate (EGCG) downregulates gelatinase-B (MMP-9) by involvement of FAK/ERK/NFκB and AP-1 in the human breast cancer cell line MDA-MB-231. Anti-Cancer Drugs. 2010;21(6):632–644. doi: 10.1097/cad.0b013e32833a4385. [DOI] [PubMed] [Google Scholar]
- 202.Milligan SA, Burke P, Coleman DT, et al. The green tea polyphenol EGCG potentiates the antiproliferative activity of c-Met and epidermal growth factor receptor inhibitors in non-small cell lung cancer cells. Clinical Cancer Research. 2009;15(15):4885–4894. doi: 10.1158/1078-0432.CCR-09-0109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Shankar S, Ganapathy S, Hingorani SR, Srivastava RK. EGCG inhibits growth, invasion, angiogenesis and metastasis of pancreatic cancer. Frontiers in Bioscience. 2008;13(2):440–452. doi: 10.2741/2691. [DOI] [PubMed] [Google Scholar]
- 204.Faderl S, Ferrajoli A, Harris D, Van Q, Priebe W, Estrov Z. WP-1034, a novel JAK-STAT inhibitor, with proapoptotic and antileukemic activity in acute myeloid leukemia (AML) Anticancer Research. 2005;25(3 B):1841–1850. [PubMed] [Google Scholar]
- 205.Shakoori A, Mai W, Miyashita K, et al. Inhibition of GSK-3β activity attenuates proliferation of human colon cancer cells in rodents. Cancer Science. 2007;98(9):1388–1393. doi: 10.1111/j.1349-7006.2007.00545.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Pizarro JG, Folch J, Esparza JL, Jordan J, Pallàs M, Camins A. A molecular study of pathways involved in the inhibition of cell proliferation in neuroblastoma B65 cells by the GSK-3 inhibitors lithium and SB-415286. Journal of Cellular and Molecular Medicine. 2009;13(9 B):3906–3917. doi: 10.1111/j.1582-4934.2008.00389.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Pallàs M, Camins A. Molecular and biochemical features in Alzheimer's disease. Current Pharmaceutical Design. 2006;12(33):4389–4408. doi: 10.2174/138161206778792967. [DOI] [PubMed] [Google Scholar]
- 208.Bhat RV, Budd Haeberlein SL, Avila J. Glycogen synthase kinase 3: a drug target for CNS therapies. Journal of Neurochemistry. 2004;89(6):1313–1317. doi: 10.1111/j.1471-4159.2004.02422.x. [DOI] [PubMed] [Google Scholar]
- 209.Cross DAE, Culbert AA, Chalmers KA, Facci L, Skaper SD, Reith AD. Selective small-molecule inhibitors of glycogen synthase kinase-3 activity protect primary neurones from death. Journal of Neurochemistry. 2001;77(1):94–102. doi: 10.1046/j.1471-4159.2001.t01-1-00251.x. [DOI] [PubMed] [Google Scholar]
- 210.Jope RS, Johnson GVW. The glamour and gloom of glycogen synthase kinase-3. Trends in Biochemical Sciences. 2004;29(2):95–102. doi: 10.1016/j.tibs.2003.12.004. [DOI] [PubMed] [Google Scholar]
- 211.Dunn N, Holmes C, Mullee M. Does lithium therapy protect against the onset of dementia? Alzheimer Disease and Associated Disorders. 2005;19(1):20–22. doi: 10.1097/01.wad.0000155068.23937.9b. [DOI] [PubMed] [Google Scholar]
- 212.Su Y, Ryder J, Li B, et al. Lithium, a common drug for bipolar disorder treatment, regulates amyloid-β precursor protein processing. Biochemistry. 2004;43(22):6899–6908. doi: 10.1021/bi035627j. [DOI] [PubMed] [Google Scholar]
- 213.Chung ES, Bok E, Sohn S, Lee YD, Baik HH, Jin BK. GT1b-induced neurotoxicity is mediated by the Akt/GSK-3/tau signaling pathway but not caspase-3 in mesencephalic dopaminergic neurons. BMC Neuroscience. 2010;11, article 74 doi: 10.1186/1471-2202-11-74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Jiang H, Guo W, Liang X, Rao Y. Both the establishment and the maintenance of neuronal polarity require active mechanisms: critical roles of GSK-3β and its upstream regulators. Cell. 2005;120(1):123–135. doi: 10.1016/j.cell.2004.12.033. [DOI] [PubMed] [Google Scholar]
- 215.Fornai F, Longone P, Cafaro L, et al. Lithium delays progression of amyotrophic lateral sclerosis. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(6):2052–2057. doi: 10.1073/pnas.0708022105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Leung E, Kim JE, Rewcastle GW, Finlay GJ, Baguley BC. Comparison of the effects of the PI3K/mTOR inhibitors NVP-BEZ235 and GSK2126458 on tamoxifen-resistant breast cancer cells. Cancer Biology and Therapy. 2011;11(11):938–946. doi: 10.4161/cbt.11.11.15527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Zhao L, Teter B, Morihara T, et al. Insulin-degrading enzyme as a downstream target of insulin receptor signaling cascade: implications for Alzheimer’s disease intervention. Journal of Neuroscience. 2004;24(49):11120–11126. doi: 10.1523/JNEUROSCI.2860-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Kim S-N, Kim S-T, Doo A-R, et al. Phosphatidylinositol 3-kinase/Akt signaling pathway mediates acupuncture-induced dopaminergic neuron protection and motor function improvement in a mouse model of parkinson's disease. International Journal of Neuroscience. 2011;121(10):562–569. doi: 10.3109/00207454.2011.591515. [DOI] [PubMed] [Google Scholar]
- 219.Van Ummersen L, Binger K, Volkman J, et al. A phase I trial of perifosine (NSC 639966) on a loading dose/maintenance dose schedule in patients with advanced cancer. Clinical Cancer Research. 2004;10(22):7450–7456. doi: 10.1158/1078-0432.CCR-03-0406. [DOI] [PubMed] [Google Scholar]
- 220.Pal SK, Reckamp K, Yu H, Figlin RA. Akt inhibitors in clinical development for the treatment of cancer. Expert Opinion on Investigational Drugs. 2010;19(11):1355–1366. doi: 10.1517/13543784.2010.520701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.(1995–2011) FDA Approved Drugs for Oncology. http://www.centerwatch.com/drug-information/fda-approvals/drug-areas.aspx?AreaID=12.
- 222.Behbakht K, Sill MW, Darcy KM, et al. Phase II trial of the mTOR inhibitor, temsirolimus and evaluation of circulating tumor cells and tumor biomarkers in persistent and recurrent epithelial ovarian and primary peritoneal malignancies. A Gynecologic Oncology Group study. Gynecologic Oncology. 2011;123(1):19–26. doi: 10.1016/j.ygyno.2011.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Oza AM, Elit L, Tsao M-S, et al. Phase II study of temsirolimus in women with recurrent or metastatic endometrial cancer: a trial of the NCIC Clinical Trials Group. Journal of Clinical Oncology. 2011;29(24):3278–3285. doi: 10.1200/JCO.2010.34.1578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Chan S, Scheulen ME, Johnston S, et al. Phase II study of temsirolimus (CCI-779), a novel inhibitor of mTOR, in heavily pretreated patients with locally advanced or metastatic breast cancer. Journal of Clinical Oncology. 2005;23(23):5314–5322. doi: 10.1200/JCO.2005.66.130. [DOI] [PubMed] [Google Scholar]
- 225.Galanis E, Buckner JC, Maurer MJ, et al. Phase II trial of temsirolimus (CCI-779) in recurrent glioblastoma multiforme: a north central cancer treatment group study. Journal of Clinical Oncology. 2005;23(23):5294–5304. doi: 10.1200/JCO.2005.23.622. [DOI] [PubMed] [Google Scholar]
- 226.Duran I, Kortmansky J, Singh D, et al. A phase II clinical and pharmacodynamic study of temsirolimus in advanced neuroendocrine carcinomas. British Journal of Cancer. 2006;95(9):1148–1154. doi: 10.1038/sj.bjc.6603419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Pandya KJ, Dahlberg S, Hidalgo M, et al. A randomized, phase II trial of two dose levels of temsirolimus (CCI-779) in patients with extensive-stage small-cell lung cancer who have responding or stable disease after induction chemotherapy: a trial of the Eastern Cooperative Oncology Group (E1500) Journal of Thoracic Oncology. 2007;2(11):1036–1041. doi: 10.1097/JTO.0b013e318155a439. [DOI] [PubMed] [Google Scholar]
- 228.Gerullis H, Ecke TH, Janusch B, et al. Long-term response in advanced bladder cancer involving the use of temsirolimus and vinflunine after platin resistance. Anti-Cancer Drugs. 2011;22(9):940–943. doi: 10.1097/CAD.0b013e328347a86a. [DOI] [PubMed] [Google Scholar]
- 229.Zardavas D, Meisel A, Samaras P, et al. Temsirolimus is highly effective as third-line treatment in chromophobe renal cell cancer. Case Reports in Oncology. 2011;4(1):16–18. doi: 10.1159/000323804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Hudes G, Carducci M, Tomczak P, et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. New England Journal of Medicine. 2007;356(22):2271–2281. doi: 10.1056/NEJMoa066838. [DOI] [PubMed] [Google Scholar]
- 231.Spilman P, Podlutskaya N, Hart MJ, et al. Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-β levels in a mouse model of alzheimer's disease. PLoS ONE. 2010;5(4, article e9979) doi: 10.1371/journal.pone.0009979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Malagelada C, Jin ZH, Jackson-Lewis V, Przedborski S, Greene LA. Rapamycin protects against neuron death in in vitro and in vivo models of Parkinson’s disease. Journal of Neuroscience. 2010;30(3):1166–1175. doi: 10.1523/JNEUROSCI.3944-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Chen S, Atkins CM, Liu CL, Alonso OF, Dietrich WD, Hu BR. Alterations in mammalian target of rapamycin signaling pathways after traumatic brain injury. Journal of Cerebral Blood Flow and Metabolism. 2007;27(5):939–949. doi: 10.1038/sj.jcbfm.9600393. [DOI] [PubMed] [Google Scholar]
- 234.Park J, Zhang J, Qiu J, et al. Combination therapy targeting Akt and mammalian target of rapamycin improves functional outcome after controlled cortical impact in mice. doi: 10.1038/jcbfm.2011.131. Journal of Cerebral Blood Flow and Metabolism. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Codeluppi S, Svensson CI, Hefferan MP, et al. The Rheb-mTOR pathway is upregulated in reactive astrocytes of the injured spinal cord. Journal of Neuroscience. 2009;29(4):1093–1104. doi: 10.1523/JNEUROSCI.4103-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Sekiguchi A, Kanno H, Ozawa H, Yamaya S, Itoi E. Rapamycin Promotes Autophagy and Reduces Neural Tissue Damage and Locomotor Impairment after Spinal Cord Injury in Mice . doi: 10.1089/neu.2011.1919. Journal of Neurotrauma. In press. [DOI] [PubMed] [Google Scholar]
- 237.Chauhan A, Sharma U, Jagannathan NR, Reeta KH, Gupta YK. Rapamycin protects against middle cerebral artery occlusion induced focal cerebral ischemia in rats. Behavioural Brain Research. 2011;225(2):603–609. doi: 10.1016/j.bbr.2011.08.035. [DOI] [PubMed] [Google Scholar]
- 238.Jimeno A, Hallur G, Chan A, et al. Development of two novel benzoylphenylurea sulfur analogues and evidence that the microtubule-associated protein tau is predictive of their activity in pancreatic cancer. Molecular Cancer Therapeutics. 2007;6(5):1509–1516. doi: 10.1158/1535-7163.MCT-06-0592. [DOI] [PubMed] [Google Scholar]
- 239.TauRx-Therapeutics-Ltd. TRx0014 in Patients With Mild or Moderate Alzheimer's Disease. ClinicalTrials.gov Identifier: NCT00515333. 2007, http://clinicaltrials.gov/ct2/show/NCT00515333?spons=%22TauRx+Therapeutics+Ltd%22&spons_ex=Y&rank=1.
- 240.Williams M. Progress in Alzheimer’s disease drug discovery: an update. Current Opinion in Investigational Drugs. 2009;10(1):23–34. [PubMed] [Google Scholar]
- 241.Zelivianski S, Spellman M, Kellerman M, et al. ERK inhibitor PD98059 enhances docetaxel-induced apoptosis of androgen-independent human prostate cancer cells. International Journal of Cancer. 2003;107(3):478–485. doi: 10.1002/ijc.11413. [DOI] [PubMed] [Google Scholar]
- 242.Yong HH, Hwa JM, You BR, et al. The MEK inhibitor PD98059 attenuates growth inhibition and death in gallic acid-treated Calu-6 lung cancer cells by preventing glutathione depletion. Molecular Medicine Reports. 2010;3(3):519–524. doi: 10.3892/mmr_00000291. [DOI] [PubMed] [Google Scholar]
- 243.Lim SJ, Lee YJ, Lee E. p38MAPK inhibitor SB203580 sensitizes human SNU-C4 colon cancer cells to exisulind-induced apoptosis. Oncology Reports. 2006;16(5):1131–1135. [PubMed] [Google Scholar]
- 244.Karunakaran S, Saeed U, Mishra M, et al. Selective activation of p38 mitogen-activated protein kinase in dopaminergic neurons of substantia nigra leads to nuclear translocation of p53 in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mice. Journal of Neuroscience. 2008;28(47):12500–12509. doi: 10.1523/JNEUROSCI.4511-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Barone FC, Irving EA, Ray AM, et al. Inhibition of p38 mitogen-activated protein kinase provides neuroprotection in cerebral focal ischemia. Medicinal Research Reviews. 2001;21(2):129–145. doi: 10.1002/1098-1128(200103)21:2<129::aid-med1003>3.0.co;2-h. [DOI] [PubMed] [Google Scholar]
- 246.Martial S, Giorgelli J-L, Renaudo A, Derijard B, Soriani O. SP600125 inhibits Kv channels through a JNK-independent pathway in cancer cells. Biochemical and Biophysical Research Communications. 2008;366(4):944–950. doi: 10.1016/j.bbrc.2007.12.027. [DOI] [PubMed] [Google Scholar]
- 247.Kim J-H, Kim TH, Kang HS, Ro J, Kim HS, Yoon S. SP600125, an inhibitor of Jnk pathway, reduces viability of relatively resistant cancer cells to doxorubicin. Biochemical and Biophysical Research Communications. 2009;387(3):450–455. doi: 10.1016/j.bbrc.2009.07.036. [DOI] [PubMed] [Google Scholar]
- 248.Wang LH, Besirli CG, Johnson EM., Jr. Mixed-lineage kinases: a target for the prevention of neurodegeneration. Annual Review of Pharmacology and Toxicology. 2004;44:451–474. doi: 10.1146/annurev.pharmtox.44.101802.121840. [DOI] [PubMed] [Google Scholar]
- 249.Leszek J, Inglot AD, Janusz M, et al. Colostrinin proline-rich polypeptide complex from ovine colostrum—a long-term study of its efficacy in Alzheimer’s disease. Medical Science Monitor. 2002;8(10):PI93–PI96. [PubMed] [Google Scholar]
- 250.Kuan CY, Burke RE. Targeting the JNK signaling pathway for stroke and parkinson’s diseases therapy. Current Drug Targets. 2005;4(1):63–67. doi: 10.2174/1568007053005145. [DOI] [PubMed] [Google Scholar]