Traumatic Brain Injury and Aging

A variety of additional factors such as age and age-associated systemic alterations can also

affect both survival and recovery from severe injury. These include changes in hormonal and

drug metabolism, nutritional status, immune function, and increased frailty, among other

factors. Given this added layer of complexity, it is not obvious that the injury itself or putative

treatments for it will behave in the same way in older subjects as they do in young adults. This

population might therefore require not only special research consideration, but also a different

treatment paradigm, which could potentially involve combination treatments designed

specifically to address an age-altered physiology.TBI is a Multiorgan Systemic Inflammatory Disorder Acute phase events with the greatest import for survival after TBI appear to be those related to inflammatory processes, especially the production of inflammatory cytokines, which is a well-recognized aspect of the physiological response to trauma.8 Levels of inflammatory cytokines are the most consistent prognostic markers of outcomes in patients with systemic inflammatory response syndrome, sepsis, multiorgan dysfunction syndrome, and multiorgan failure,9 all of which are commonly the proximate cause of death after CNS trauma 7,10,11 and generally result from an imbalance between pro- and anti-inflammatory responses to severe injury.10 We therefore support the view that TBI should be considered a systemic and not just a focal problem, especially in the context of inflammation and short-term survival.Systemic effects are also evident in response to oxidative stress. Shohami et al.12 reported that TBI induces a cascade of highly reactive oxygen species that can damage brain tissue and other organs throughout the body. Using sophisticated methods to measure the ability of tissue to scavenge free radicals as a measure of oxidative stress, the investigators found that closed head injury in male rats led to widespread changes in brain, liver, lung, skin, kidney, and intestine within 24 h of the insult. More recently Moinard et al.13 found significantly altered metabolism and mitochondrial activity in the liver triggered by the TBI-induced inflammatory cascade in the brain. Histological examination of liver tissue showed that the TBI caused immune cell

infiltration, edema, fibrosis, and necrosis that helped to block normal liver metabolism and

homeostasis.

Inflammatory markers are not just causally related to mortality after TBI, but also appear to

be a good index of the extent of injury 9,14 and reliable independent predictors of injury

outcome.15–18 Although the levels of the cytokines tumor necrosis factor-α (TNF α) and

interleukin-1β (IL-1β) increase after severe injury, it is the level of IL-6 that is considered the

most accurate prognostically, because it is the chief regulator of acute hepatic response 19 and

correlates with systemic inflammation and outcome.9 There is also evidence that early

increased levels of IL-6 can be a marker of high risk of complication and organ failure.20

Although it has been argued that systemic inflammation is so complex that it defies traditional

reductionist definitions and may not be useful in predicting system behaviors,21 modulating

the production of inflammatory cytokines during the acute phase could be of benefit in reducing

mortality and improving the prognosis for better functional outcomes. For example, there is

recent evidence of sex differences in patients with multiple trauma, with premenopausal

women showing significantly lower plasma cytokines and less multiorgan failure and sepsis,

compared with age-matched men.22 It is possible that these sex differences in response to injury

are attributable to higher circulating levels of progesterone (P 4) in female subjects,23,24 an

intriguing hypothesis, given the success of P 4 in treating TBI.

Progesterone and Brain Injury Treatment

A number of recent publications have demonstrated effectiveness of P 4 treatment in

experimental models of TBI.25–29 It has been consistently shown that post-injury

administration of P 4 can attenuate the cytological, morphological, and functional deficits

NIH-PA Author Manuscript

NIH-PA Author Manuscript

NIH-PA Author Manuscript

caused by traumatic injury.27,30–35 Unlike other sex steroids, P 4 is synthe-sized by

oligodendrocytes in the brain, where it then acts on membrane-bound and classical nuclear

steroid receptors.36 Progesterone can also affect water channel proteins (aquaporins) to

modulate both vasogenic and cytotoxic edema,37,38 reduce glutamate toxicity, mediate toxic

calcium influx by its antagonist effects on sigma receptors, upregulate GABA A receptors to

reduce excitotoxic damage, and reduce apoptosis and necrosis.29,36 It is likely that the

pleiotropic and systemic nature of P 4 is responsible for its effectiveness in the treatment of

TBI. As one recent review suggests, “given the multifactorial nature of the secondary injury

process, it is unlikely that targeting a single factor will result in significant improvement in

outcome. Conversely, targeting several injury factors may be the most likely therapeutic

approach to improve outcome ”4 (p. 29, emphasis added). Progesterone is just such a

multifactorial agent.

The preclinical findings on neuroprotective effects of P 4 have been confirmed in humans in a

recently completed phase IIa single-center clinical trial for P 4 in the treatment of moderate to

severe adult TBI (N = 100).39 Mortality among patients given P 4 intravenously for 3 days after

the injury was less than half that of controls provided standard-of-practice care but no hormone

(13.6% vs 30.4%). Functional outcomes at 30 days were significantly better for moderately

injured patients given P 4 than for the placebo group, and a data safety monitoring board

appointed by the U.S. National Institutes of Health found no serious adverse events attributable

to P 4 treatment. This was the first successful clinical trial for the treatment of TBI in more than

30 years of research.

A second independent, randomized double-blind study from China examined P 4 in 159 patients

with severe TBI given an intramuscular course of P 4 treatment over 5 days. Similar beneficial

outcomes on morbidity and mortality were observed at both 30 days and 6 months after injury,

again without any serious adverse events caused by the treatment.40

A phase III, 17-center, double-blind, randomized clinical trial for TBI will start enrolling about

1000 patients in January 2010 (NCT00822900; https://www.360docs.net/doc/5018902203.html,).

One specific reason for increased survival in P 4-treated TBI patients may be the ability of P 4to substantially reduce systemic acute-phase inflammation and so prevent multiorgan

dysfunction syndrome and multiorgan failure. In support of this idea, Chen et al.41 found that

contusion injury to the cerebral cortex in rats induced expression of IL-1β and TNF α in the

intestinal mucosa, followed by apoptosis in the intestinal mucosa. A 5-day course of treatment

with P 4 reduced both the cytokines and cell death in the intestine, demonstrating the systemic

benefits of P 4 administration.Aging and Brain Injury

Although the incidence of TBI has decreased in most age groups because of improved primary

prevention (such as seat belt and safety helmet use), it has increased by 21% in people over

the age of 65.42 Age is itself an independent predictor of mortality due to TBI, which in the

geriatric population is twice that of younger age groups.43 The age distribution of

hospitalizations due to TBI has the most pronounced peak late in the eighth decade,44 and the

highest mortality rates from TBI, ranging from 60.9% to 86.8% depending on the study, occur

in people 75 to 80 years old.45 Given the magnitude of the problem, it is important to address

the effectiveness of any treatment specifically in aged subjects.

Old animals are fatter, more sedentary and irritable, and (like most old people) have sensory,

cognitive, and motor deficits that can exacerbate injury-induced impairments and impede

functional recovery. Old rats and old people also metabolize drugs differently than young ones,

so doses and agents that work in younger subjects might not work in older animals, or could

NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

even aggravate the injury. A number of systemic physiological changes associated with the

aging process could also alter drug effectiveness, such as altered metabolism and endogenous

hormonal milieu,46–50 impaired immune function,51–53 increased levels of systemic

inflammation,54 and reduced physiological reserve or frailty.55,56 Additional changes within

the CNS, including reduced plasticity 57 and more subtle chemical, structural, morphological,

and network alterations,58,59 can also significantly affect the ability of an older animal or person

to survive and recover from severe injury.

CNS effects of aging

Although it is established that there is no overt loss of neurons in the brain with normal aging,

a number of more subtle structural, chemical, and metabolic changes occur,58,59 both at the

level of individual neurons and in medium-scale neuronal networks, that can significantly

affect the ability of the CNS to adapt to internal and environmental changes. Other changes in

the CNS in response to aging are similar to the changes that occur in other cells, and include

increased oxidative stress, altered protein accumulation, nucleic acid damage, and dysfunction

of energy homeostasis.59 The cellular changes during normal aging make neurons increasingly

susceptible to excitotoxic damage 59 through the impairment of ion pumps,60 dysregulation of

Ca 2+ homeostasis,59 and decreased mitochondrial function.61 Given that all these processes

are involved in the evolution of the injury after traumatic insult, the process of aging itself

could significantly increase vulnerability and impair the potential for recovery from TBI in

aged individuals.

There is evidence that the brains of aged animals exhibit gene expression profiles characteristic

of microglial activation and neuroinflammation.62–65 Microglia themselves also appear to

respond to stimulation with an amplified inflammatory response in aged animals.66 This

neuroinflammatory priming can significantly affect the development of neurological

dysfunction. Under normal conditions these inflammatory changes are temporary, and the

microglia return to a dormant state as soon as the immune challenge is resolved. Aging,

however, creates a brain environment in which microglial sensitivity to immune activation is

increased and microglial activation does not resolve, which can lead to the pathogenesis of

neurological disease.66 This situation is likely to exacerbate secondary injury in older subjects

after TBI, in that trauma can lead not only to a severe systemic immune response,67 but also

to immune factor release from the damaged cells within the brain itself.68

Exaggerated neuroinflammation can also interfere with neuroplasticity during the recovery

phase, and inflammatory cytokines such as TNF α, IL-1β, and IL-6 appear to directly interfere

with long-term potentiation,69–71 memory consolidation,72 neurite outgrowth,73 and

hippocampal neurogenesis.74 These data suggest that continued presence of reactive microglia

in the aged brain creates an environment permissive to a prolonged and amplified

neuroinflammatory response that can lead to subsequent complications, especially after TBI.66Systemic effects of aging and immunosenescence

In addition to alterations in the CNS, age-related changes in the immune system are also

common and have been implicated in virtually all age-associated disease processes.53 Aging

is associated with a general activation of the inflammatory response, which, because of the

chronic antigenic stress on innate immunity experienced over a lifetime, becomes the basis for

the onset of inflammatory diseases 51 and an increasing inability to mount an appropriate

immune response to antigenic stimulation.53 With aging there is also a decrease in the

production of anti-inflammatory hormones,75 as well as a general tendency toward production

of elevated amounts of proinflammatory cytokines by peripheral blood mononuclear cells.54

NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

The fulminating inflammatory state associated with increasing age has been dubbed

inflammaging .54 This condition is systemic but also has specific effects within the CNS.66

Given the importance of inflammatory cytokines such as TNF α, IL-1β, and IL-6 in both

behavioral modulation and the evolution of traumatic injury, it is likely that an increased

neuroinflammatory cytokine response in the elderly patient can disrupt neuronal synaptic

plasticity, establishing a CNS environment predisposed to long-lasting complications, as well

as a reduced ability to recover from trauma.66 In fact, the aged brain appears to exist in a chronic

state of inflammation that is associated with increased immune reactivity and continuous low-

level production of central inflammatory cytokines.68

Endocrine effects of aging

In addition to structural and immune decline, human aging is associated with decreased activity

in a number of systemic hormones, including thyroid hormone,47 sex steroids,46,49,76 growth

hormone,48 insulin-like growth factor type 1 (IGF-I),48 and 25-hydroxyvitamin D 3. 77,50,78

Especially intriguing is the relationship between endocrine decline and the change in

immunological function. Some research suggests that proinflammatory cytokines may

downregulate the physiological responses to a variety of hormones, including insulin, growth

hormone, IGF-I, thyroid hormone, and estrogens.79

TNF α, IL-1, and IL-6 can specifically modulate the release of growth hormone, growth

hormone releasing hormone, and somatostatin,80 and are known to reduce concentrations of

IGF-I 81 and increase the concentrations of glucocorticoids 82 in the serum of human patients.

Lower serum IGF-I and dehydroepiandrosterone sulfate levels were found in frail (compared

with nonfrail) elderly individuals,56 and an inverse correlation between serum IL-6 and IGF-

I was noted only in frail individuals.56 Elevated levels of inflammatory cytokines 83–86 and the

development of frailty in older adults 87,88 have also been associated with vitamin D deficiency

(D-deficiency).

These data can be taken to suggest that the immune system and its age-associated dysfunction

may be intimately related to the functioning of the neuroendocrine system, and that this

relationship may further contribute to the development of systemic insufficiency and

vulnerability to injury.

Aging and vitamin D deficiency

Of all the endocrine and nutritional disruptions that affect the elderly population, D-deficiency

has recently received considerable attention, partly because an estimated 1 billion people

worldwide exhibit D-deficiency or insufficiency.89 In the northeastern United States, studies

show a prevalence from 32% in healthy adults 90 to approximately 50% for adolescents and

preadolescents.91–93 The most dramatic statistics for D-deficiency, however, come from

studies in the elderly population, which show that 40 to 100% of community-dwelling

American and European older men and women are D-deficient. Even higher averages are seen

in the ill and institutionalized.94–102

Vitamin D-deficiency has been associated with many systemic disorders, including infectious,

inflammatory, and autoimmune conditions,103–109 cardiovascular disease,110,111 hypertension

and atherosclerosis,112 neuromuscular function,113 cancer,114 neurodegenerative diseases,115,116 and neuropsychological and functional outcomes in the elderly population.117 In fact,

D-deficiency appears to be correlated with many if not most of the problems associated with

advanced age, especially those with an inflammatory component.103,106 There is also evidence

that the level of serum vitamin D is a key marker of frailty,83,88 and that it is associated with

elevated levels of IL-6.118NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

It is worth noting here that vitamin D is in fact not a vitamin, but rather a secosteroid hormone

with the same cholesterol backbone as other steroids (including P 4) and with its own class of

nuclear steroid receptors and signaling mechanisms.119,120 In its biologically active form, 1,25-

dihydroxyvitamin D 3 (vitamin D hormone, or VDH) is a hormone with sites of action

throughout the CNS; it can be considered a neuroactive steroid, because it is both synthesized

and has actions in the nervous system. Vitamin D hormone is also a known and potent

modulator of the cell cycle, immune function, and calcium homeostasis. As such, it may be in

important compound not only as an endogenous hormone, but also as a treatment in its own

right, especially in combination with P 4.Aging, Brain Injury, and Neurosteroids Progesterone in aged rats Given the promising results with progesterone in young adult animals, we sought to determine if it would work as well in aged animals.121 The working hypothesis in our aging studies was that P 4 would be effective in treating TBI in aged rats. We measured levels of inflammatory proteins, cell death, edema, and behavior during the acute phase of injury (24–72 h after TBI)in aged rats (20 months old, approximately equivalent to 60 years in humans) to determine the potential of P 4 in reducing mortality after TBI. Injured animals treated with 8 mg/kg and 16mg/kg P 4 beginning within the first hour after surgery showed decreased expression of cyclooxygenase-2, IL-6, and nuclear factor κB (NF κB) at all time points examined, indicating a reduction in the acute inflammatory process compared with the aged rats given vehicle. The 16 mg/kg P 4 group also showed reduced neuronal apoptosis at all time points, and decreased edema and improved locomotor outcomes. Although the lowest P 4 dosage used in previous studies in younger animals (8 mg/kg) was effective in the aged animals, overall it was not as effective as 16 mg/kg, suggesting potentially altered P 4 kinetics and metabolism in the older animals.We also observed an association between increased levels of P-glycoprotein and reduced

edema, suggesting that P 4 may reduce cerebral edema both through its antiinflammatory effects

on cytokine levels and through direct effects on the integrity of the blood–brain barrier. P-

glycoprotein is regulated through the pregnane X receptor (PXR)122,123 and plays a critical

role in removing toxic products from the cell. Progesterone has been shown to exert

neuroprotective effects through the PXR.124 This raises the intriguing possibility that some of

the post-TBI benefits of P 4 may be effected through this relatively novel signaling mechanism.

Consequences of D-deficiency for TBI and P 4 treatment

Given the effectiveness of P 4 in aged rats after TBI and the prevalence of D-deficiency in the

elderly population, we asked whether D-deficiency would affect the outcome of a brain injury

and whether it would interfere with the benefits of P 4 treatment in aged rats. Based on the

current literature, we hypothesized that D-deficiency would exacerbate inflammation and

reduce or eliminate the benefits of P 4 treatment after TBI in aged animals. Because early onset

of inflammation is taken as a reliable prognostic indicator of mortality in human patients with

significant trauma, we measured acute inflammatory proteins, cell death, DNA damage, and

short-term behavior as indicators of inflammation and secondary damage in D-deficient aged

animals after TBI.

In general, we observed increased inflammation in aged rats with D-deficiency, whether our

subjects were uninjured, injured but untreated, or injured and treated with P 4.125 Our results

confirmed previous studies 103,106,126 suggesting that D-deficiency establishes a higher

baseline level of inflammation, in effect priming the system for an increased immune–

inflammatory response after brain injury. This elevated acute-phase response correlated with

NIH-PA Author Manuscript

NIH-PA Author Manuscript

NIH-PA Author Manuscript

increased cell death and DNA damage, indicating a more severe secondary injury process.

Vitamin D deficiency also affected sickness behaviors, such as movement and grooming, which

strongly correlated (by general linear models) with the expression of TNF α and IL-6.

Our data can be taken to indicate that short-term alterations in locomotor behavior could serve

as a rough behavioral indicator of brain inflammation in the acute phase after injury. Our

models also indicated that both TNF α and IL-6 were significantly increased by D-deficiency

and that both cytokines showed deficiency–treatment interaction effects. We take this to mean

that the increased expression of these inflammatory cytokines may play an important role in

the cascade of toxic mechanisms underlying the attenuation of P 4 benefits in D-deficiency.

Also notable is the fact that, although most of the variability in TNF α was accounted for by

injury, the level of IL-6 was affected primarily by D-deficiency. This fits with other

independent data connecting IL-6 levels with D-deficiency, frailty, and inflammation 88,118,

127,128 and suggests to us that IL-6 may be the key cytokine involved in the detrimental effects

of D-deficiency. The elevation in IL-6 was most evident in our data comparing D-deficient

and D-normal animals after TBI and P 4 treatment. Most of the other cytokines were elevated

in D-deficient animals twofold or threefold, but IL-6 was increased nearly fivefold by 72 h

after injury.

Management of VDH deficiency

Given the data from human studies showing that the level of IL-6 is the most accurate

prognostic indicator of survival in the acute phase after TBI, our findings could have significant

implications in the clinical setting. We suggest that elderly people be screened for potential D-

deficiency and provided with supplementation therapy after TBI, because they could be more

frail and thus more likely to die from their injuries. Powner et al.129 point out that, during

rehabilitation from severe TBI, as many as 40% of patients suffer from endocrine dysfunctions

associated with sickness behaviors such as chronic fatigue, decreased libido, amenorrhea,

increased anxiety, memory failure, depression, and anorexia, among others. Although for some

of these symptoms it is speculative that vitamin D could be a helpful treatment, the costs of

vitamin D supplementation are minor and it is easy to give, and the implications for the

treatment of a number of disease conditions could be important for clinical practice.

Although it seems reasonable that supplementation with vitamin D in a deficient state would

be equivalent to ongoing vitamin D sufficiency (i.e., acute correction of deficiency should be

equivalent to no deficiency), there is no a priori reason to make this assumption. At 24 h after

injury, we observed no differences in inflammatory cytokines between VDH-treated deficient

and D-normal animals. By 72 h after injury, while there was still no difference with TNF α and

IL-1β was still lower than in D-normal animals, levels of IL-6, NF κB p65, and cell death were

all higher in VDH-treated deficient than in D-normal animals. These findings can be taken to

suggest that, at least with reference to IL-6 levels, acutely correcting D-deficiency is not as

good as maintaining D-normal status. In other words, prevention could be better than acute

treatment.

Vitamin D deficiency as a public health issue has received much attention recently in the

popular press, as well as in the medical literature. Now we can add another benefit to

maintaining a normal vitamin D status: it is not just good for the arteries and protective against

cancer, it can also be a primary intervention against TBI—in some ways, the equivalent of

wearing a seatbelt. Although further research needs to be done on this issue, we suggest that

the findings are likely to apply across the developmental spectrum and not just to the elderly

population (where it might be exacerbated because of complications from other health-related

factors).NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

Combining P 4 With Vdh In Treating TBI In The Aged Population

Rationale for combinatorial therapy in treating TBI

Combination therapy is somewhat new to the area of brain injury, but it is a well-established pharmacological approach to a number of other diseases, such as HIV–AIDS or tuberculosis,although most drug development still focuses on individual drugs targeting one or at most a few specific mechanisms. Two issues are involved: drug combination and target promiscuity,or pleiotropy .Rationales for combining drugs include targeting multiple divergent mechanisms and overcoming single-drug limitations such as receptor kinetics, pharmacology, and signaling pathways.130,131 This idea has recently gained ground in the treatment of TBI.5 Because both P 4 and VDH are pleiotropic and multifunctional, it is difficult to predict specific mechanisms of interaction, but both could be used to target cell death after TBI, for example, as one of the many mechanisms by which a brain injury evolves. On the one hand, P 4 would reduce cell death by preventing release of cytochrome c from the mitochondria, upregulating the antiapoptotic Bcl-2 protein, and downregulating the proapoptotic Bax protein.1,26,132 Vitamin D hormone, on the other hand, would prevent the reactivation of cell cycle machinery,133–135 which is a common step toward apoptosis in terminally differentiated neurons,136,137 and would upregulate nerve growth factor (NGF), providing a strong external prosurvival signal.138,139 The two compounds would thus act in synergy and provide benefits greater than either compound https://www.360docs.net/doc/5018902203.html,bining P 4 and VDH, pleiotropic drugs that work primarily through intracellular receptors and transcription factors likewise merits serious consideration, as meeting the criteria posited by Vink and Nimmo 4 in their review of multifunctional drugs for head injury. We strongly agree that a combinatorial approach to treatment is not only reasonable but may be essential,given the complexity and heterogeneity of human TBI and the fact that, of the 130 preclinical

monotherapy drugs that have shown promise in treating brain injury, all have failed when taken

to clinical trial.5

Effectiveness of P 4–VDH combination after TBI in aged rats

We recently demonstrated effectiveness of a combination of P 4 and VDH in both in vitro and

in vivo models. Combining VDH and P 4 was shown to be effective in vitro in increasing the

survival of primary cortical neurons under glutamate challenge, with the combination showing

more neuroprotection than either compound alone at best dose.140 We applied the same

treatment concept to D-deficient aged rats with TBI and showed that the only treatment that

reduced proteins measured (TNF α, IL-1β, IL-6, NF κB p65, activated caspase-3, and p53) in

all cases by 72 h after injury was the combination of P 4 and VDH (5 mg/kg in a single dose)

compared with vehicle or either compound given alone. The combination treatment was also

the only one that dramatically improved behavioral parameters, which our statistical models

again showed to be strongly correlated with systemic inflammation and levels of TNF α and

IL-6. Should these results be translatable to humans, the clinical implications are obvious: a

combination treatment of P 4 and VDH given to elderly patients with TBI should improve

survival over P 4 given alone to the same population.

Briefly summarized, we conclude that 1) in aged rats with brain injury, P 4 is effective in

reducing acute inflammation, a key indicator of survival in human patients; 2) D-deficiency

increases acute-phase inflammation and attenuates the benefits of P 4 treatment in aged rats

with TBI, suggesting that such a deficiency could increase mortality after brain injury in human

patients; 3) a combination of P 4 and VDH partially reverses the effects of D-deficiency and

reduces post-TBI acute inflammation in aged rats, suggesting that, should these results translate

NIH-PA Author Manuscript

NIH-PA Author Manuscript

NIH-PA Author Manuscript

to the clinic, a simple vitamin D injection combined with P 4 could help save many human lives.

Vink and Nimmo 4 address this theme directly:

The brain is the most complex organ in the body, and significant gaps still exist in

our understanding of normal brain function and how it is affected by acute injury.

Unlike other organ systems, we cannot readily relate cellular function to the function

of the organ as a whole.… Nonetheless, we do understand that acute brain injury is a

heterogeneous type of injury, made up of immediate and delayed anatomic, molecular,

biochemical and physiological events that are both complex and multi-faceted. So

complex are these interactions that the concept of a single magic bullet is no longer

accepted by the research community, and the focus has now turned to interventions

that can modulate a number of independent injury factors simultaneously [p. 37].

Potential mechanisms of action

Although P 4 acts through a number of mechanisms to reduce cell death and improve functional

recovery after TBI,36,141 here we propose that a key process in its ability to reduce

inflammation after injury is the ability to inhibit proinflammatory Th1 and also stimulate anti-

inflammatory Th2 immune responses. Although the Th1/Th2 paradigm is not unproblematic,142 it is useful in understanding the role of the adaptive immune system in inflammation. Our

explanatory model is based on the fact that Th1/Th2 differentiation marks a decisive event very

much like a phase transition in the development of extended inflammation after severe injury.

As suggested earlier, this reduction in systemic inflammation may be one of the key

mechanisms by which P 4 significantly increases survival after TBI in human patients.39,40

With age, the general response of the immune system moves toward a proinflammatory

condition 54,66 that is associated with frailty and the increased susceptibility of aged organisms

to external insult.55,56,143 This fact of increased inflammation and decreased immune

flexibility with age could explain why we needed a larger dose of P 4 in older animals to obtain

similar effects on acute-phase reactants. In other words, anti-inflammatory effects need to be

maximized to overcome the underlying predisposition toward inflammatory hyper-reactivity.

Under conditions of D-deficiency, the proinflammatory Th1 bias is elevated to a point at which

P 4 alone cannot counteract the amplification of the inflammatory cascade after injury. One

way to prevent overwhelming inflammation after injury, then, is to combine pleiotropic drugs

such as P 4 and VDH. Although the various gene regulatory and signaling mechanisms will be

different, they will presumably synergize at the systemic level to inhibit Th1 differentiation,

reduce the acute phase, and potentially improve outcome and survival.Conclusion

A general approach to conceptualizing aging views it as a loss of systemic integration and

adaptability that leads to a reduced ability to resist environmental perturbation, eventually

leading to death.144 This decline of complex functioning in aging and disease has been observed

in various systems, from cardiac rhythms and gait to cerebral autoregulation and large-scale

brain network integration.145–151 Because of its association with frailty and various other age-

related diseases, D-deficiency appears to have a similar destabilizing effect.83,152 Progesterone

and especially VDH can be viewed as system stabilizers, as is evident from their beneficial

effects on processes that range from DNA stability and cell cycle control to systemic immune

function and hormone production.50,133,153 Because severe TBI is essentially a massive

perturbation that leads to activation and likely extreme overactivation of defensive responses

(as in the case of inflammation, which although beneficial in the short term can also be highly

damaging when overactive or prolonged), the introduction of stabilizing factors may allow for

a reduction of this early damage and an improved ability to recover.

NIH-PA Author Manuscript NIH-PA Author Manuscript

NIH-PA Author Manuscript

Acknowledgments

Portions of the research done at Emory University and described in this review were supported by National Institutes

of Health grants 5R01NS048451 and 5R01HD061971. The authors would like to thank Leslie Mc-Cann for editorial

assistance.

References 1. Schumacher M, Guennoun R, Stein DG, De Nicola AF. Progesterone: therapeutic opportunities for neuroprotection and myelin repair. Pharmacol Ther 2007;116:77–107. [PubMed: 17659348]2. Thurman DJ, Alverson C, Dunn KA, Guerrero J, Sniezek JE. Traumatic brain injury in the United States: a public health perspective. J Head Trauma Rehabil 1999;14:602–615. [PubMed: 10671706]3. Povlishock JT, Katz DI. Update of neuropathology and neurological recovery after traumatic brain injury. J Head Trauma Rehabil 2005;20:76–94. [PubMed: 15668572]4. Vink R, Nimmo AJ. Multifunctional drugs for head injury. Neurotherapeutics 2009;6:28–42. [PubMed:19110197]5. Margulies S, Hicks R, Combination Therapies for Traumatic Brain Injury Workshop https://www.360docs.net/doc/5018902203.html,bination therapies for traumatic brain injury: prospective considerations. J Neurotrauma 2009;26:925–939. [PubMed: 19331514]6. Narayan RK, Michel ME, Ansell B, et al. Clinical trials in head injury. J Neurotrauma 2002;19:503–557. [PubMed: 12042091]7. Zygun DA, Kortbeek JB, Fick GH, Laupland KB, Doig CJ. Non-neurologic organ dysfunction in severe traumatic brain injury. Crit Care Med 2005;33:654–660. [PubMed: 15753760]8. Harwood PJ, Giannoudis PV, van Griensven M, Krettek C, Pape HC. Alterations in the systemic inflammatory response after early total care and damage control procedures for femoral shaft fracture in severely injured patients. J Trauma 2005;58:446–452. discussion 452–444. [PubMed: 15761335]9. Pape HC, Tsukamoto T, Kobbe P, Tarkin I, Katsoulis S, Peitzman A. Assessment of the clinical course with inflammatory parameters. Injury 2007;38:1358–1364. [PubMed: 18048038]10. Keel M, Trentz O. Pathophysiology of polytrauma. Injury 2005;36:691–709. [PubMed: 15910820]

11. Zygun D. Non-neurological organ dysfunction in neurocritical care: impact on outcome and

etiological considerations. Curr Opin Crit Care 2005;11:139–143. [PubMed: 15758594]

12. Shohami E, Gati I, Beit-Yannai E, Trembovler V, Kohen R. Closed head injury in the rat induces

whole body oxidative stress: overall reducing antioxidant profile. J Neurotrauma 1999;16:365–376.

[PubMed: 10369557]

13. Moinard C, Gupta S, Besson V, et al. Evidence for impairment of hepatic energy homeostasis in head-

injured rat. J Neurotrauma 2008;25:124–129. [PubMed: 18260795]

14. Bochicchio GV, Napolitano LM, Joshi M, McCarter RJ Jr, Scalea TM. Systemic inflammatory

response syndrome score at admission independently predicts infection in blunt trauma patients. J

Trauma 2001;50:817–820. [PubMed: 11379594]

15. Malone DL, Kuhls D, Napolitano LM, McCarter R, Scalea T. Back to basics: validation of the

admission systemic inflammatory response syndrome score in predicting outcome in trauma. J

Trauma 2001;51:458–463. [PubMed: 11535891]

16. Napolitano LM, Ferrer T, McCarter RJ Jr, Scalea TM. Systemic inflammatory response syndrome

score at admission independently predicts mortality and length of stay in trauma patients. J Trauma

2000;49:647–652. discussion 652–643. [PubMed: 11038081]

17. Nast-Kolb D, Waydhas C, Gippner-Steppert C, et al. Indicators of the posttraumatic inflammatory

response correlate with organ failure in patients with multiple injuries. J Trauma 1997;42:446–454.

discussion 454–445. [PubMed: 9095112]

18. Nuytinck HK, Offermans XJ, Kubat K, Goris JA. Whole-body inflammation in trauma patients: an

autopsy study. Arch Surg 1988;123:1519–1524. [PubMed: 3056336]

19. Gabay C, Kushner I. Acute-phase proteins and other systemic responses to inflammation. N Engl J

Med 1999;340:448–454. [PubMed: 9971870]

NIH-PA Author Manuscript

NIH-PA Author Manuscript

NIH-PA Author Manuscript

20. Gebhard F, Pfetsch H, Steinbach G, Strecker W, Kinzl L, Brückner UB. Is interleukin 6 an early marker of injury severity following major trauma in humans? Arch Surg 2000;135:291–295.[PubMed: 10722030]21. Vodovotz Y, Csete M, Bartels J, Chang S, An G. Translational systems biology of inflammation.PLoS Comput Biol 2008;4:1–6. e1000014.22. Frink M, Pape HC, van Griensven M, Krettek C, Chaudry IH, Hildebrand F. Influence of sex and age on MODS and cytokines after multiple injuries. Shock 2007;27:151–156. [PubMed: 17224789]23. Attella MJ, Nattinville A, Stein DG. Hormonal state affects recovery from frontal cortex lesions in adult female rats. Behav Neural Biol 1987;48:352–367. [PubMed: 3689284]24. Roof RL, Stein DG. Progesterone treatment attenuates brain edema following contusion injury in male and female rats. Restor Neurol Neurosci 1992;4:425–427.25. Goss CW, Hoffman SW, Stein DG. Behavioral effects and anatomic correlates after brain injury: a progesterone dose-response study. Pharmacol Biochem Behav 2003;76:231–242. [PubMed:14592674]26. Djebaili M, Guo Q, Pettus EH, Hoffman SW, Stein DG. The neurosteroids progesterone and allopregnanolone reduce cell death, gliosis, and functional deficits after traumatic brain injury in rats.J Neurotrauma 2005;22:106–118. [PubMed: 15665606]27. Pettus EH, Wright DW, Stein DG, Hoffman SW. Progesterone treatment inhibits the inflammatory agents that accompany traumatic brain injury. Brain Res 2005;1049:112–119. [PubMed: 15932748]28. Djebaili M, Hoffman SW, Stein DG. Allopregnanolone and progesterone decrease cell death and cognitive deficits after a contusion of the rat pre-frontal cortex. Neuroscience 2004;123:349–359.[PubMed: 14698743]29. Stein DG. Progesterone exerts neuroprotective effects after brain injury. Brain Res Rev 2008;57:386–397. [PubMed: 17826842]30. Guo Q, Baronne L, Hoffman SW, Guennoun R, Stein DG. Progesterone's effects on aquaporin-4expression after traumatic brain injury in male rats. J Neurotrauma 2004;21:1303. abstract.31. Shear DA, Galani R, Hoffman SW, Stein DG. Progesterone protects against necrotic damage and behavioral abnormalities caused by traumatic brain injury. Exp Neurol 2002;178:59–67. [PubMed:

12460608]

32. McCullough LD, Hurn PD. Estrogen and ischemic neuroprotection: an integrated view. Trends

Endocrinol Metab 2003;14:228–235. [PubMed: 12826329]

33. Stein DG. Brain damage, sex hormones and recovery: a new role for progesterone and estrogen?

Trends Neurosci 2001;24:386–391. [PubMed: 11410269]

34. Stein DG, Hoffman SW. Concepts of CNS plasticity in the context of brain damage and repair. J Head

Trauma Rehabil 2003;18:317–341. [PubMed: 16222128]

35. Stein DG, Hoffman SW. Estrogen and progesterone as neuropro-tective agents in the treatment of

acute brain injuries. Pediatr Rehabil 2003;6:13–22. [PubMed: 12745891]

36. Schumacher M, Guennoun R, Ghoumari A, et al. Novel perspectives for progesterone in hormone

replacement therapy, with special reference to the nervous system. Endocr Rev 2007;28:387–439.

[PubMed: 17431228]

37. Guo Q, Sayeed I, Baronne LM, Hoffman SW, Guennoun R, Stein DG. Progesterone administration

modulates AQP4 expression and edema after traumatic brain injury in male rats. Exp Neurol 2006;198:469–478. [PubMed: 16445913]

38. Sayeed I, Guo Q, Hoffman SW, Stein DG. Allopregnanolone, a progesterone metabolite, is more

effective than progesterone in reducing cortical infarct volume after transient middle cerebral artery occlusion. Ann Emerg Med 2006;47:381–389. [PubMed: 16546625]

39. Wright DW, Kellermann AL, Hertzberg VS, et al. ProTECT: a randomized clinical trial of

progesterone for acute traumatic brain injury. Ann Emerg Med 2007;49:391–402. [PubMed:17011666]

40. Xiao G, Wei J, Yan W, Wang W, Lu Z. Improved outcomes from the administration of progesterone

for patients with acute severe traumatic brain injury: a randomized controlled trial. Crit Care 2008;12:R61. [PubMed: 18447940]

NIH-PA Author Manuscript

NIH-PA Author Manuscript

NIH-PA Author Manuscript

41. Chen G, Shi JX, Qi M, Wang HX, Hang CH. Effects of progesterone on intestinal inflammatory response, mucosa structure alterations, and apoptosis following traumatic brain injury in male rats.J Surg Res 2008;147:92–98. [PubMed: 17868700]42. Adekoya N, Thurman DJ, White DD, Webb KW. Surveillance for traumatic brain injury deaths:United States, 1989–1998. MMWR Surveill Summ 2002;51:1–14. [PubMed: 12529087]43. Mosenthal AC, Lavery RF, Addis M, et al. Isolated traumatic brain injury: age is an independent predictor of mortality and early outcome. J Trauma 2002;52:907–911. [PubMed: 11988658]44. McArthur DL, Chute DJ, Villablanca JP. Moderate and severe traumatic brain injury: epidemiologic,imaging and neuropathologic perspectives. Brain Pathol 2004;14:185–194. [PubMed: 15193031]45. Gómez PA, Lobato RD, Boto GR, De la Lama A, González PJ, de la Cruz J. Age and outcome after severe head injury. Acta Neurochir (Wien) 2000;142:373–380. discussion 380–381. [PubMed:10883332]46. Vermeulen A, Kaufman JM, Goemaere S, van Pottelberg I. Estradiol in elderly men. Aging Male 2002;5:98–102. [PubMed: 12198740]47. Vermeulen A, Kaufman JM. Ageing of the hypothalamo–pituitary–testicular axis in men. Horm Res 1995;43:25–28. [PubMed: 7721258]48. Lanfranco F, Gianotti L, Giordano R, Pellegrino M, Maccario M, Arvat E. Ageing, growth hormone and physical performance. J Endocrinol Invest 2003;26:861–872. [PubMed: 14964439]49. Araujo AB, O'Donnell AB, Brambilla DJ, et al. Prevalence and incidence of androgen deficiency in middle-aged and older men: estimates from the Massachusetts Male Aging Study. J Clin Endocrinol Metab 2004;89:5920–5926. [PubMed: 15579737]50. Rammos G, Tseke P, Ziakka S. Vitamin D, the renin–angiotensin system, and insulin resistance. Int Urol Nephrol 2008;40:419–426. [PubMed: 18193490]51. Vasto S, Candore G, Balistreri CR, et al. Inflammatory networks in ageing, age-related diseases and longevity. Mech Ageing Dev 2007;128:83–91. [PubMed: 17118425]52. Gruver AL, Hudson LL, Sempowski GD. Immunosenescence of ageing. J Pathol 2007;211:144–156.[PubMed: 17200946]53. Bulati M, Pellicanò M, Vasto S, Colonna-Romano G. Understanding ageing: biomedical and

bioengineering approaches, the immunologic view. Immun Ageing 2008;5:9. [PubMed: 18768084]

54. Franceschi C, Capri M, Monti D, et al. Inflammaging and anti-inflammaging: a systemic perspective

on aging and longevity emerged from studies in humans. Mech Ageing Dev 2007;128:92–105.

[PubMed: 17116321]

55. Walston J, Hadley EC, Ferrucci L, et al. Research agenda for frailty in older adults: toward a better

understanding of physiology and etiology: summary from the American Geriatrics Society/National Institute on Aging Research Conference on Frailty in Older Adults. J Am Geriatr Soc 2006;54:991–1001. [PubMed: 16776798]

56. Fried LP, Tangen CM, Walston J, et al. Frailty in older adults: evidence for a phenotype. J Gerontol

A Biol Sci Med Sci 2001;56:M146–M156. [PubMed: 11253156]

57. Hof PR, Morrison JH. The aging brain: morphomolecular senescence of cortical circuits. Trends

Neurosci 2004;27:607–613. [PubMed: 15374672]

58. Dickstein DL, Kabaso D, Rocher AB, Luebke JI, Wearne SL, Hof PR. Changes in the structural

complexity of the aged brain. Aging Cell 2007;6:275–284. [PubMed: 17465981]

59. Mattson MP, Magnus T. Ageing and neuronal vulnerability. Nat Rev Neurosci 2006;7:278–294.

[PubMed: 16552414]

60. Mattson MP. Excitotoxic and excitoprotective mechanisms: abundant targets for the prevention and

treatment of neurodegenerative disorders. Neuromolecular Med 2003;3:65–94. [PubMed: 12728191]

61. Melov S. Modeling mitochondrial function in aging neurons. Trends Neurosci 2004;27:601–606.

[PubMed: 15374671]

62. Godbout JP, Chen J, Abraham J, et al. Exaggerated neuroinflammation and sickness behavior in aged

mice following activation of the peripheral innate immune system. FASEB J 2005;19:1329–1331.

[PubMed: 15919760]

NIH-PA Author Manuscript

NIH-PA Author Manuscript

NIH-PA Author Manuscript

63. Frank MG, Barrientos RM, Biedenkapp JC, Rudy JW, Watkins LR, Maier SF. mRNA up-regulation of MHC II and pivotal pro-inflammatory genes in normal brain aging. Neurobiol Aging 2006;27:717–722. [PubMed: 15890435]64. Perry VH, Matyszak MK, Fearn S. Altered antigen expression of microglia in the aged rodent CNS.Glia 1993;7:60–67. [PubMed: 8423063]65. Sheffield LG, Berman NE. Microglial expression of MHC class II increases in normal aging of nonhuman primates. Neurobiol Aging 1998;19:47–55. [PubMed: 9562503]66. Godbout JP, Johnson RW. Age and neuroinflammation: a lifetime of psychoneuroimmune consequences. Neurol Clin 2006;24:521–538. [PubMed: 16877122]67. Lenz A, Franklin GA, Cheadle WG. Systemic inflammation after trauma. Injury 2007;38:1336–1345.[PubMed: 18048040]68. Dilger RN, Johnson RW. Aging, microglial cell priming, and the discordant central inflammatory response to signals from the peripheral immune system. J Leukoc Biol 2008;84:932–939. [PubMed:18495785]69. Griffin R, Nally R, Nolan Y, McCartney Y, Linden J, Lynch MA. The age-related attenuation in long-term potentiation is associated with microglial activation. J Neurochem 2006;99:1263–1272.[PubMed: 16981890]70. Li AJ, Katafuchi T, Oda S, Hori T, Oomura Y. Interleukin-6 inhibits long-term potentiation in rat hippocampal slices. Brain Res 1997;748:30–38. [PubMed: 9067442]71. Vereker E, O'Donnell E, Lynch MA. The inhibitory effect of interleukin-1β on long-term potentiation is coupled with increased activity of stress-activated protein kinases. J Neurosci 2000;20:6811–6819.[PubMed: 10995825]72. Barrientos RM, Higgins EA, Biedenkapp JC, et al. Peripheral infection and aging interact to impair hippocampal memory consolidation. Neurobiol Aging 2006;27:723–732. [PubMed: 15893410]73. Neumann H, Schweigreiter R, Yamashita T, Rosenkranz K, Wekerle H, Barde YA. Tumor necrosis factor inhibits neurite outgrowth and branching of hippocampal neurons by a rho-dependent mechanism. J Neurosci 2002;22:854–862. [PubMed: 11826115]74. Vallières L, Campbell IL, Gage FH, Sawchenko PE. Reduced hippocampal neurogenesis in adult

transgenic mice with chronic astrocytic production of interleukin-6. J Neurosci 2002;22:486–492.

[PubMed: 11784794]

75. Straub RH, Miller LE, Sch?lmerich J, Zietz B. Cytokines and hormones as possible links between

endocrinosenescence and immunosenescence. J Neuroimmunol 2000;109:10–15. [PubMed:

10969175]

76. Jiang M, Huhtaniemi I. Polymorphisms in androgen and estrogen receptor genes: effects on male

aging. Exp Gerontol 2004;39:1603–1611. [PubMed: 15582276]

77. Elmadfa I, Meyer AL. Body composition, changing physiological functions and nutrient requirements

of the elderly. Ann Nutr Metab 2008;52:2–5. [PubMed: 18382069]

78. Holick MF. Sunlight and vitamin D for bone health and prevention of autoimmune diseases, cancers,

and cardiovascular disease. Am J Clin Nutr 2004;80(6 Suppl):1678S–1688S. [PubMed: 15585788]

79. Ferrucci L, Ble A, Bandinelli S, Lauretani F, Suthers K, Guralnik JM. A flame burning within. Aging

Clin Exp Res 2004;16:240–243. [PubMed: 15462468]

80. Talhouk RS, Saadé NE, Mouneimne G, Masaad CA, Safieh-Garabedian B. Growth hormone releasing

hormone reverses endotoxin-induced localized inflammatory hyperalgesia without reducing the upregulated cytokines, nerve growth factor and gelatinase activity. Prog Neuropsychopharmacol Biol Psychiatry 2004;28:625–631. [PubMed: 15276687]

81. Priego T, Granado M, Ibá?ez de Cáceres I, Martín AI, Villanúa MA, López-Calderón A. Endotoxin

at low doses stimulates pituitary GH whereas it decreases IGF-I and IGF-binding protein-3 in rats.J Endocrinol 2003;179:107–117. [PubMed: 14529571]

82. Beishuizen A, Thijs LG. Endotoxin and the hypothalamo–pituitary–adrenal (HPA) axis. J Endotoxin

Res 2003;9:3–24. [PubMed: 12691614]

83. Topinkova E. Aging, disability and frailty. Ann Nutr Metab 2008;52:6–11. [PubMed: 18382070]

84. Nagpal S, Na S, Rathnachalam R. Noncalcemic actions of vitamin D receptor ligands. Endocr Rev

2005;26:662–687. [PubMed: 15798098]

NIH-PA Author Manuscript

NIH-PA Author Manuscript

NIH-PA Author Manuscript

85. Richards JB, Valdes AM, Gardner JP, et al. Higher serum vitamin D concentrations are associated with longer leukocyte telomere length in women. Am J Clin Nutr 2007;86:1420–1425. [PubMed:17991655]86. Moore ME, Piazza A, McCartney Y, Lynch MA. Evidence that vitamin D 3 reverses age-related inflammatory changes in the rat hippocampus. Biochem Soc Trans 2005;33:573–577. [PubMed:16042547]87. Boxer RS, Dauser DA, Walsh SJ, Hager WD, Kenny AM. The association between vitamin D and inflammation with the 6-minute walk and frailty in patients with heart failure. J Am Geriatr Soc 2008;56:454–461. [PubMed: 18194227]88. Morley JE, Haren MT, Rolland Y, Kim MJ. Frailty. Med Clin North Am 2006;90:837–847. [PubMed:16962845]89. Holick MF. Vitamin D deficiency. N Engl J Med 2007;357:266–281. [PubMed: 17634462]90. Tangpricha V, Pearce EN, Chen TC, Holick MF. Vitamin D insufficiency among free-living healthy young adults. Am J Med 2002;112:659–662. [PubMed: 12034416]91. Gordon CM, DePeter KC, Feldman HA, Grace E, Emans SJ. Prevalence of vitamin D deficiency among healthy adolescents. Arch Pediatr Adolesc Med 2004;158:531–537. [PubMed: 15184215]92. Nadziejko C, Fang K, Narciso S, et al. Effect of particulate and gaseous pollutants on spontaneous arrhythmias in aged rats. Inhal Toxicol 2004;16:373–380. [PubMed: 15204753]93. Sullivan SS, Rosen CJ, Halteman WA, Chen TC, Holick MF. Adolescent girls in Maine are at risk for vitamin D insufficiency. J Am Diet Assoc 2005;105:971–974. [PubMed: 15942551]94. Chatfield SM, Brand C, Ebeling PR, Russell DM. Vitamin D deficiency in general medical inpatients in summer and winter. Intern Med J 2007;37:377–382. [PubMed: 17535381]95. Corino A, D'Amelio P, Gancia R, et al. Hypovitaminosis D in internal medicine inpatients. Calcif Tissue Int 2007;80:76–80. [PubMed: 17308988]96. Eyles D, Brown J, Mackay-Sim A, McGrath J, Feron F. Vitamin D 3 and brain development.Neuroscience 2003;118:641–653. [PubMed: 12710973]97. Eyles DW, Smith S, Kinobe R, Hewison M, McGrath JJ. Distribution of the vitamin D receptor and 1 α-hydroxylase in human brain. J Chem Neuroanat 2005;29:21–30. [PubMed: 15589699]

98. Hewison M, Zehnder D, Bland R, Stewart PM. 1α-Hydroxylase and the action of vitamin D. J Mol

Endocrinol 2000;25:141–148. [PubMed: 11013342]

99. Holick MF. McCollum Award Lecture, 1994. Vitamin D: new horizons for the 21st century. Am J

Clin Nutr 1994;60:619–630. [PubMed: 8092101]

100. Larrosa M, Gratacòs J, Vaqueiro M, Prat M, Campos F, Roqué M. Prevalence of hypovitaminosis

D in elderly institutionalized residents: influence of a substitutive treatment [In Spanish]. Med Clin (Barc) 2001;117:611–614. [PubMed: 11714466]

101. McGrath JJ, Féron FP, Burne TH, Mackay-Sim A, Eyles DW. Vitamin D 3: implications for brain

development. J Steroid Biochem Mol Biol 2004:89–90. 557–560.

102. Townsend K, Evans KN, Campbell MJ, Colston KW, Adams JS, Hewison M. Biological actions of

extra-renal 25-hydroxyvitamin D-1α-hydroxylase and implications for chemoprevention and treatment. J Steroid Biochem Mol Biol 2005;97:103–109. [PubMed: 16081283]

103. Hayes CE, Nashold FE, Spach KM, Pedersen LB. The immunological functions of the vitamin D

endocrine system. Cell Mol Biol (Noisy-le-grand) 2003;49:277–300. [PubMed: 12887108]

104. DeLuca HF, Zierold C. Mechanisms and functions of vitamin D. Nutr Rev 1998;56:S4–10.

discussion S54–75. [PubMed: 9564171]

105. Griffin MD, Xing N, Kumar R. Vitamin D and its analogs as regulators of immune activation and

antigen presentation. Annu Rev Nutr 2003;23:117–145. [PubMed: 12651965]

106. Cantorna MT, Zhu Y, Froicu M, Wittke A. Vitamin D status, 1,25-dihydroxyvitamin D 3, and the

immune system. Am J Clin Nutr 2004;80:1717S–1720S. [PubMed: 15585793]

107. Mahon BD, Wittke A, Weaver V, Cantorna MT. The targets of vitamin D depend on the

differentiation and activation status of CD4 positive T cells. J Cell Biochem 2003;89:922–932.

[PubMed: 12874827]

108. Holick MF. Evolution and function of vitamin D. Recent Results Cancer Res 2003;164:3–28.

[PubMed: 12899511]

NIH-PA Author Manuscript

NIH-PA Author Manuscript

NIH-PA Author Manuscript

109. Holick MF. Vitamin D: a millenium perspective. J Cell Biochem 2003;88:296–307. [PubMed:12520530]110. Melamed ML, Muntner P, Michos ED, et al. Serum 25-hydroxyvitamin D levels and the prevalence of peripheral arterial disease: results from NHANES 2001 to 2004. Arterioscler Thromb Vasc Biol 2008;28:1179–1185. [PubMed: 18417640]111. Martins D, Wolf M, Pan D, et al. Prevalence of cardiovascular risk factors and the serum levels of 25-hydroxyvitamin D in the United States: data from the Third National Health and Nutrition Examination Survey. Arch Intern Med 2007;167:1159–1165. [PubMed: 17563024]112. Onyszchuk G, He YY, Berman NE, Brooks WM. Detrimental effects of aging on outcome from traumatic brain injury: a behavioral, magnetic resonance imaging, and histological study in mice.J Neurotrauma 2008;25:153–171. [PubMed: 18260798]113. Pfeifer M, Begerow B, Minne HW. Vitamin D and muscle function. Osteoporos Int 2002;13:187–194. [PubMed: 11991436]114. Peterlik M, Cross HS. Vitamin D and calcium deficits predispose for multiple chronic diseases. Eur J Clin Invest 2005;35:290–304. [PubMed: 15860041]115. Garcion E, Wion-Barbot N, Montero-Menei CN, Berger F, Wion D. New clues about vitamin D functions in the nervous system. Trends Endocrinol Metab 2002;13:100–105. [PubMed: 11893522]116. Kalueff AV, Lou YR, Laaksi I, Tuohimaa P. Increased anxiety in mice lacking vitamin D receptor gene. Neuroreport 2004;15:1271–1274. [PubMed: 15167547]117. Dawson-Hughes B. Serum 25-hydroxyvitamin D and functional outcomes in the elderly. Am J Clin Nutr 2008;88:537S–540S. [PubMed: 18689397]118. McCarty MF. Secondary hyperparathyroidism promotes the acute phase response: a rationale for supplemental vitamin D in prevention of vascular events in the elderly. Med Hypotheses 2005;64:1022–1026. [PubMed: 15780504]119. Dusso AS, Brown AJ, Slatopolsky E. Vitamin D. Am J Physiol Renal Physiol 2005;289:F8–F28.[PubMed: 15951480]120. van Etten E, Mathieu C. Immunoregulation by 1,25-dihydroxyvitamin D 3: basic concepts. J Steroid Biochem Mol Biol 2005;97:93–101. [PubMed: 16046118]

121. Cutler SM, Cekic M, Miller DM, Wali B, VanLandingham JW, Stein DG. Progesterone improves

acute recovery after traumatic brain injury in the aged rat. J Neurotrauma 2007;24:1475–1486.

[PubMed: 17892409]

122. Bauer B, Hartz AM, Fricker G, Miller DS. Pregnane X receptor up-regulation of P-glycoprotein

expression and transport function at the blood–brain barrier. Mol Pharmacol 2004;66:413–419.

[PubMed: 15322232]

123. Miller DS, Bauer B, Hartz AM. Modulation of P-glycoprotein at the blood–brain barrier:

opportunities to improve central nervous system pharmacotherapy. Pharmacol Rev 2008;60:196–209. [PubMed: 18560012]

124. Langmade SJ, Gale SE, Frolov A, et al. Pregnane X receptor (PXR) activation: a mechanism for

neuroprotection in a mouse model of Niemann-Pick C disease. Proc Natl Acad Sci U S A

2006;103:13807–13812. [PubMed: 16940355]

125. Cekic M, Cutler SM, Vanlandingham JW, Stein DG. Vitamin D deficiency reduces the benefits of

progesterone treatment after brain injury in aged rats. Neurobiol Aging. 2009 May 29; Epub ahead of print.

126. Lips P. Vitamin D physiology. Prog Biophys Mol Biol 2006;92:4–8. [PubMed: 16563471]

127. Miller RR, Hicks GE, Shardell MD, et al. Association of serum vitamin D levels with inflammatory

response following hip fracture: the Baltimore Hip Studies. J Gerontol A Biol Sci Med Sci

2007;62:1402–1406. [PubMed: 18166692]

128. Van den Berghe G, Van Roosbroeck D, Vanhove P, Wouters PJ, De Pourcq L, Bouillon R. Bone

turnover in prolonged critical illness: effect of vitamin D. J Clin Endocrinol Metab 2003;88:4623–4632. [PubMed: 14557432]

129. Powner DJ, Boccalandro C, Alp MS, Vollmer DG. Endocrine failure after traumatic brain injury in

adults. Neurocrit Care 2006;5:61–70. [PubMed: 16960299]

130. Faden AI. Neuroprotection and traumatic brain injury: the search continues. Arch Neurol

2001;58:1553–1555. [PubMed: 11594912]

NIH-PA Author Manuscript

NIH-PA Author Manuscript

NIH-PA Author Manuscript

131. Gingrich MB, Traynelis SF. Serine proteases and brain damage: is there a link? Trends Neurosci 2000;23:399–407. [PubMed: 10941185]132. Brinton RD, Thompson RF, Foy MR, et al. Progesterone receptors: form and function in brain. Front Neuroendocrinol 2008;29:313–339. [PubMed: 18374402]133. Chatterjee M. Vitamin D and genomic stability. Mutat Res 2001;475:69–87. [PubMed: 11295155]134. Cozzolino M, Lu Y, Finch J, Slatopolsky E, Dusso AS. p21WAF1 and TGF-α mediate parathyroid growth arrest by vitamin D and high calcium. Kidney Int 2001;60:2109–2117. [PubMed: 11737585]135. Jensen SS, Madsen MW, Lukas J, Binderup L, Bartek J. Inhibitory effects of 1α,25-dihydroxyvitamin D 3 on the G 1-S phase-controlling machinery. Mol Endocrinol 2001;15:1370–1380. [PubMed: 11463860]136. Cernak I, Stoica B, Byrnes KR, Di Giovanni S, Faden AI. Role of the cell cycle in the pathobiology of central nervous system trauma. Cell Cycle 2005;4:1286–1293. [PubMed: 16082214]137. 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. Proc Natl Acad Sci U S A 2005;102:8333–8338. [PubMed: 15923260]138. Brown J, Bianco JI, McGrath JJ, Eyles DW. 1,25-dihydroxyvitamin D 3 induces nerve growth factor,promotes neurite outgrowth and inhibits mitosis in embryonic rat hippocampal neurons. Neurosci Lett 2003;343:139–143. [PubMed: 12759183]139. Wion D, MacGrogan D, Neveu I, Jehan F, Houlgatte R, Brachet P. 1,25-Dihydroxyvitamin D 3 is a potent inducer of nerve growth factor synthesis. J Neurosci Res 1991;28:110–114. [PubMed:1904101]140. Atif F, Sayeed I, Ishrat T, Stein DG. Progesterone with vitamin D affords better neuroprotection against excitotoxicity in cultured cortical neurons than progesterone alone. Mol Med 2009;15:328–336. [PubMed: 19603099]141. Stein DG, Wright DW, Kellermann AL. Does progesterone have neuroprotective properties? Ann Emerg Med 2008;51:164–172. [PubMed: 17588708]142. Kidd P. Th1/Th2 balance: the hypothesis, its limitations, and implications for health and disease.Altern Med Rev 2003;8:223–246. [PubMed: 12946237]

143. Paganelli R, Di Iorio A, Cherubini A, et al. Frailty of older age: the role of the endocrine–immune

interaction. Curr Pharm Des 2006;12:3147–3159. [PubMed: 16918440]

144. Lipsitz LA. Physiological complexity, aging, and the path to frailty. Sci Aging Knowledge Environ

2004;2004:pe16. [PubMed: 15103055]

145. Lipsitz LA, Goldberger AL. Loss of ‘complexity’ and aging: potential applications of fractals and

chaos theory to senescence. JAMA 1992;267:1806–1809. [PubMed: 1482430]

146. Goldberger AL, Peng CK, Lipsitz LA. What is physiologic complexity and how does it change with

aging and disease? Neurobiol Aging 2002;23:23–26. [PubMed: 11755014]

147. Schulte-Frohlinde V, Ashkenazy Y, Goldberger AL, et al. Complex patterns of abnormal heartbeats.

Phys Rev E Stat Nonlin Soft Matter Phys 2002;66(3 Pt 1):031901. [PubMed: 12366146]

148. Costa M, Goldberger AL. Multiscale entropy analysis of complex physiologic time series. Phys Rev

Lett 2002;89(6):068102. [PubMed: 12190613]

149. Hausdorff JM, Peng CK, Goldberger AL, Stoll AL. Gait unsteadiness and fall risk in two affective

disorders: a preliminary study. BMC Psychiatry 2004;4:39. [PubMed: 15563372]

150. Novak V, Yang AC, Lepicovsky L, Goldberger AL, Lipsitz LA, Peng CK. Multimodal pressure-

flow method to assess dynamics of cerebral autoregulation in stroke and hypertension. Biomed Eng Online 2004;3:39. [PubMed: 15504235]

151. Andrews-Hanna JR, Snyder AZ, Vincent JL, et al. Disruption of large-scale brain systems in

advanced aging. Neuron 2007;56:924–935. [PubMed: 18054866]

152. Villoslada P, Steinman L, Baranzini SE. Systems biology and its application to the understanding

of neurological diseases. Ann Neurol 2009;65:124–139. [PubMed: 19260029]

153. Adorini L, Penna G. Control of autoimmune diseases by the vitamin D endocrine system. Nat Clin

Pract Rheumatol 2008;4:404–412. [PubMed: 18594491]

NIH-PA Author Manuscript

NIH-PA Author Manuscript

NIH-PA Author Manuscript

密室逃脱计划书

密室逃脱计划书 一、项目介绍 真人密室逃脱,打破了电脑游戏的局限和束缚,原汁原味的展现了密室的精髓,让玩家能过通过自己的双眼和双手,经过逻辑思考和观察力,不断的发现线索和提示,最重要的是团队的合作,能够顺利的逃脱。整个过程充满了未知性和不确定性,在紧张的场景氛围中,真正的融入到故事背景中去,这是电脑游戏所无法提供的乐趣。 真人密室也是在2011年才在国内正式起步,相较于国外已经成熟的体系,还未完全被大众所接受。2011年9月20日,第一家主营真人密室逃脱的俱乐部,在杭州正式成立。据可靠统计,全国目前已经有超过500家以上真人密室逃脱俱乐部,其中以北上广三地发展最为迅速。福州地区2013年3月左右,开始出现第一家真人密室逃脱俱乐部。我们的密室逃脱俱乐部名为八度空间,于2013年5月开始经营,属于福州地区比较早的密室商家。 二、市场分析 1.市场前景分析: 我们都是生活在平静生活中的普通人,每天重复着相同的事情,毫无生机。即便有着看电影、唱K、泡吧等种种休闲活动,但是对于追求新鲜的年轻人来说,闲暇时间还是显得越来越乏味。而要脱离这种提不起劲的困境,逐渐兴起的真人主题密室逃脱无疑会是一个最好的选择。密室逃脱实体游戏项目,既好玩刺激,又可以让每个体验者在游戏中充分调用智慧和体力,享受酐畅淋漓的畅快感觉。 据调查,福州2010年人口711万,现在福州的密室逃脱有6家(有实力),一年的订单数为22100个,票价35左右。和福州的土地面积差不多大小的洛阳和北京,与之比较,洛阳和北京的密室逃脱比福州早一年开始,2010年洛阳人口680万,现在洛阳一年的订单数为27200个,但是只有一家的密室逃脱(有实力),为寡头市场。2012年北京人口2069万,现在北京一年的订单数为49000个,票价70元,11家密室逃脱。福州与洛阳比较:店铺越多,接收客源面积越大,订单数越多。所以福州的未来市场的需求量为增幅状态。福州与北京比较:价钱越贵,订单数越少。所以福州的未来市场的需求量为增幅状态。 注:以上数据来源于美团网和中国知网,店铺收入不单单来源于网络的团购,还有其他收入,所以这只是代表部分的收入,所以当做消极悲观者的眼光来看待市场的需求量也是为增幅状态。 作为一个新兴行业,它的市场是还没有饱和的,你的质量比别家的好,有吸引力,客源量就会增加。结论:2014年起,至少一年,市场的需求量为增幅状态。 2.与传统娱乐项目对比的优势 ①体验刺激 或许过山车的尖叫只是生理上的一种释放,但是主题密室逃脱却能够从心理上让体验者感受刺激。各种精心布置的主题场景,暗含玄机的种种物品,还有与场景相配的音乐,这些都能够让体验者产生一种身临其境的感觉,从而可以更快地融入密室逃脱当中。当然,情节悬疑也是好玩的密室逃脱中一个重要影响因素。只有环环相扣的故事情节以及层层迭进的线索,才能够带动体验者更充分地享受推理和逃脱的过程。 ②高科技力挺 密室逃脱要做得好仅仅依靠故事情节和环境布置就太单薄了,高科技产品的使用也是使密室逃脱更加真实的一个诀窍。运用高科技物理机械和电子幻灯制造悬疑的场景气氛,炫酷的室内特技效果让场景显得更加逼真,而红外线、夜视仪等道具的使用也充分调动了体验者的紧张感和积极性。这些高科技的使用让密室逃脱不仅仅只是一个游戏项目,还成为一场科技产品展示的体验与经历。 ③价值无限 如果只将其当作是一个娱乐项目未免有点太低看真人密室逃脱了,因为在整个的逃脱过程当中,除了让体验者感受刺激和悬疑以外,还考验了每个体验者的智力、耐性以及良好的团队协作力。只有将团队中每个人找到的线索结合起来,才能够找到正确的出路。当然,这中间也需要有人指挥,有人配合,一旦团

小游戏----密室逃脱

活动名称:大班数学探索活动----密室逃脱 设计思路: 偶然的机会听孩子们分享自己玩游戏的经验,发现他们对比较有挑战性的游戏如密室逃脱闯关等非常感兴趣,于是,我就想:能不能用游戏这样一个载体,将数学这一学科的某些知识呈现在里面?于是,我就按照自己的理解,把生活中常见的物品分成几个层次:将游戏材料有的散点状放置,有的向不同方向有序排列,有的重叠排列等,投放到科学探索区内,观察幼儿的游戏情况。在幼儿操作材料过程中,我发现他们数数时的方法、速度、能力等都有异同,因此,我梳理了幼儿的一些数数经验,结合大班幼儿的年龄特点,生成了本次活动。 活动目标:1、在幼儿观察、比较、推理中,尝试通过操作材料,探索可以将物品数正确的方法。 2、仔细倾听同伴的想法,乐意分享观察方式。 活动准备:幼儿操作卡(每人2张)、PPT4张、笔、夹子、照片。 活动过程: 1、呈现密室,激发幼儿兴趣:了解幼儿数数的能力。 出示PPT1,谈话引出课题 1、讲解密室逃脱游戏规则。 师:今天,老师带你们玩密室逃脱,你知道这个游戏怎么玩吗?师讲解游戏规则:密室逃脱就是我们进入房间,通过数房间里各种物品的数量来推算出房门的密码,从而打开房门,逃出密室。 2、报数游戏:让幼儿数数接龙。 师:清点人数完毕,请你们摆一个最漂亮、最帅气的姿势跟老师们打个招呼,不然,待会出不来可能没有人来救你。 3、谈话:你能数到几? 师:刚才集体报数完全正确,现在,我问问小朋友,你最多能数到几?我说一个数字,你接着往下数,往下数()个数。(教师一方面关注幼儿数数是否正确,另一方面关注幼儿从十位数数到百位数数的能力,同时,观察其他幼儿,聆听每个幼儿的数数情况。)师:小朋友们都能数到很大的数,真了不起!但你们数数的正确性怎么样呢?老师会在游戏中检查你们的本领。 2、进入密室:探索正确数数的方法。 第一环节:来到密室一:出示PPT2 师:我们已经进入密室,现在,赶紧来找找怎打开房门的密码。(引导幼儿找到门上的密码锁,通过寻找密码打开锁从而逃出密室。)密码锁提示我们找哪些物品? 1、幼儿完成相应操作卡,正确数出PPT上相框、靠枕、猫及红花的数量。 师:密码锁提示我们:要仔细找找密室里有几幅相框?靠枕有几个?猫有几只?红花有几朵?把你观察到的结果记录在对应的空格里,不会写数字的用圆点代替。(教师重点关注猫和红花的记录情况) 2、幼儿交流自己的观察结果。教师及时提升幼儿表达的数数要领: A、一起告诉我,相框有几幅?靠枕有几个?师总结:东西比较少,数起来很方便; B、数猫可就不简单了,请您告诉我,你数到有几只猫?你在哪里找到了第×只猫?师总结:要把东西数清楚是需要讲究方法,要从上到下、从左到右、从前到后,只要按照一定的顺序就能数清楚。(教师要重点关注幼儿的表达,如方位是否正确,语句是否完整,还要关注幼儿数猫的方法) C、这里的花特别多,你是怎么数的?你是怎么数的?黄花能不能数进去?引导幼儿说出2个2个一起数的规律。师总结:数东西的时候,不但要讲究顺序,而且要仔细看清楚题目,

密室逃脱相关 (2)

Q1:别人做密室逃脱 从大二时,开始接触密室逃脱游戏,到今年寒假一天能玩上三、四次,“既然大家都喜欢,干嘛不自己也开一个呢?”黄皓月介绍,决定开一个“密室逃脱”店,很偶然,也很仓促。5个爱玩密室的女孩当即一拍即合,其中有两个女生已经工作,三个在校生中,黄皓月也即将本科毕业,她还“跨界”选修了金融专业的双学位。 三月份,考察江汉路所有密室逃脱店、玩遍40多个主题,敲定店址; 四月份,店面装修,自己设计主题、自己安装机关、自己制作装置……; “五一”,新店开张,首日客流量过百,不大的客厅里,挤满了前来“尝鲜”的同龄人。 开业的前一天,女孩们都在店里通宵加班,“创业比考研费精力多了,四月份的时候,天天熬夜。”黄皓月告诉记者,开店,比想象中的复杂、琐碎。对于“学霸”的称号,她觉得那是上高中以前的事了,从高二起,一直到现在,她更愿意把自己定义为“学霸班上的玩货”。 中考前,黄皓月因成绩优秀,提前保送到当地最好的学校——新洲一中“火箭班”。然而高一的第一次月考,成绩仅排在班上十几名,随后几次月考,更是滑落到二十几名。“我已经很努力了,学习成绩就是上不去。”家长的不理解,老师的不重视,让曾经的“神童公主”的骄傲光环瞬间破碎,从台上最闪亮的“明星”,跌落成台下普通观众,巨大的落差,让她一度“放松”自己,按照自己的意愿学习、生活,成为学霸班上的“另类”。 “其实二十几名已经很好了,至少可以考上武大华科,但那个时候没有人告诉我,我以为自己真的很差。”说到这里的时候,黄皓月的眼中有一丝伤感和落寞,虽然之后成绩时常徘徊在班上倒数几名,但高考总分依然超过一本分数线,考入湖北大学生物科学专业。

小游戏----密室逃脱

活动名称: 大班数学探索活动----密室逃脱 设计思路: 偶然的机会听孩子们分享自己玩游戏的经验,发现他们对比较有挑战性的游戏如密室逃脱闯关等非常感兴趣,于是,我就想: 能不能用游戏这样一个载体,将数学这一学科的某些知识呈现在里面?于是,我就按照自己的理解,把生活中常见的物品分成几个层次: 将游戏材料有的散点状放置,有的向不同方向有序排列,有的重叠排列等,投放到科学探索区内,观察幼儿的游戏情况。在幼儿操作材料过程中,我发现他们数数时的方法、速度、能力等都有异同,因此,我梳理了幼儿的一些数数经验,结合大班幼儿的年龄特点,生成了本次活动。 活动目标: 1、在幼儿观察、比较、推理中,尝试通过操作材料,探索可以将物品数正确的方法。 2、仔细倾听同伴的想法,乐意分享观察方式。 活动准备: 幼儿操作卡(每人2张)、PPT4张、笔、夹子、照片。 活动过程: 1、呈现密室,激发幼儿兴趣: 了解幼儿数数的能力。 出示PPT1,谈话引出课题 1、讲解密室逃脱游戏规则。 师:

今天,老师带你们玩密室逃脱,你知道这个游戏怎么玩吗?师讲解游戏规则: 密室逃脱就是我们进入房间,通过数房间里各种物品的数量来推算出房门的密码,从而打开房门,逃出密室。 2、报数游戏: 让幼儿数数接龙。 师: 清点人数完毕,请你们摆一个最漂亮、最帅气的姿势跟老师们打个招呼,不然,待会出不来可能没有人来救你。 3、谈话: 你能数到几? 师: 刚才集体报数完全正确,现在,我问问小朋友,你最多能数到几?我说一个数字,你接着往下数,往下数()个数。(教师一方面关注幼儿数数是否正确,另一方面关注幼儿从十位数数到百位数数的能力,同时,观察其他幼儿,聆听每个幼儿的数数情况。)师: 小朋友们都能数到很大的数,真了不起!但你们数数的正确性怎么样呢?老师会在游戏中检查你们的本领。 2、进入密室: 探索正确数数的方法。 第一环节: 来到密室一: 出示PPT2

《密室逃脱》碧绿色房间闯关攻略详解

《密室逃脱》碧绿色房间闯关攻略详解 继博士的家之后,小编接下来要为大家介绍密室逃脱碧绿色房间闯关攻略。密室逃脱,又 叫TAKAGISM ,是一款密室闯关游戏,游戏需要玩游戏者在游戏中寻找线索,一步一步地走出 密室。《密室逃脱》游戏的玩法很简单,只需用鼠标即可。整个游戏过程都是在一间房子里进行的,游戏的主人公不知道为什么被困在这里(每集都交待了不同的原因,但这些原因都是相当的 离奇)T akagism的逃离房间系列是非常经典的解迷小游戏,自从作者推出第一个版本Crimson Room后便深受世界各地网友的喜爱,并因此掀起了一股“逃离房间热”。本文将详细介绍密室逃脱碧绿色房间如何闯关。 1、开灯,左转(点画面的左下角,右转同理)在鞋橱的鞋下边发现“若”字图片;鞋橱底发现镜框。 2、左转,在土黄色的垫子底下发现“般”字图片。 3、左转,在壁橱的最上一格中左右边分别发现“多”字图片和一束香。二格的红笔记本有锁打 不开,在最下面的小橱里发现黄影集里两张照片。 4、点地上的红毯子,发现骷髅!点击靠近,在左手手腕处发现钥匙,右手发现药瓶,点小腿处的红绳物体可以把毯子叠起,并发现脚上有密码锁(暂时不开),点物品栏中的红毯,再点下边的“查看物体”(即"About Item"),可放大,点大图的红毯子得到十字架状物体。 百度攻略&口袋巴士提供 1

5、左转,在斜对着蓝门处,(就是那个有窟窿的蓝门,不是一开始的那个门)。点头顶的 灯罩,在灯罩上发现“罗”字图片(小红块,仔细点),在墙上发现“ 蜜”字图片(小白块)。 6、左转,在小冰箱里发现“ 波”字图片和一罐啤酒(啤酒先不要拿,因为骷髅要冰冻的)。在旁边的红桶里发现张纸,打开是些黑头发。记住:点装黑头发的纸,然后点击查看物体,放大纸,多点几下纸,然后就可以在红桶底下得到打火机。 7、用钥匙开红笔记本打开,在日记中可以找到女孩的生日,那就是开骷髅脚上的密码。(通常会是在日记的第四页,里面有一句话“The day after tomorrow is her birthday”,这一天的简单写法 就是密码。你先看你所翻到的那页日记上是几月几号,再在那个日期后加两天,就是小女孩子的 生日,那一天的简写就是密码。(如9月10日,简写就是0910,这就是密码),记下密码。继续翻日记,翻到最后得到一张CD。 8、放大装有头发的纸,点打火机,再点纸,可以烧出东西来,放大药瓶,里面有两粒药,一 张写有箴言的纸。放大绿十字,按照箴言的顺序位置和烧出来得数字相对应(形位相同),把字( 6张图片)填在十字架状物体上(位置随机的),放对会变成个四方体,放下暂不用。 9、在壁橱的第3格,有香炉和磬(旁边红的是槌),把装有CD的CD盒放在香炉后面,把香放香炉左,罐啤放香左(要冰镇的啊,冰一两分钟就可以拿出来了,先把啤酒从冰箱拿出来,点显 百度攻略&口袋巴士提供 2

真人密室逃脱指南

现在,非常流行真人密室逃脱的游戏,让你进入密室里面,想尽一切办法逃出来。很多人不明白这个怎么玩,小编今日以自己的经验为你介绍。 方法/步骤 1首先,你在网上查一下在你所在的地方,有什么真人的密室。最好去的人比较多,年龄是不受限制的,但是尽量不要带16岁以下的,带去了没用,而且又花费了一张票钱。 2真人密室有很多主题。比如,诅咒;医院;梦游等等。还有很多电影的,比如,生化危机;电锯惊魂;盗墓,都是很火热的主题。所谓,主题,就是说密室里面的道具、背景、风格是哪种。 3选好主题之后。价格方面,其实所以密室都是差不多的价格,除非它的规模很大。价格基本在40到65一人,团购价,如果是选择在网上团购,那么要先打电话预约时间。注意:如果你明天要去,最好是提前半天打电话。这种真人游戏需要预约。 4打电话联系,会定下你们的时间,要求是提前15分钟到她们店里。如果你不选择网上团购,那么就到现场再付费,但是也是需要预约的。因为,真人密室非常受人追捧。而且,每场游戏时间都是一个小时。 5一定要提前去。去了之后,店内都有桌游可以随便玩,有饮料供应。所以也不必担心无聊。去之前最好少带随身物品,女士包包,都要少带。进去之前,工作人员会让你交出手机(有干扰),放在保险箱里。大可放心。 6进去之后,会有纸张写密室的故事背景。进去之后,一定要注意看周围的东西。一般来说,密室里面大多都是机械化的东西,像空调就不用找了,有纸张会提示你们什么东西不用去找。然后你们就要去找东西,也就是线索。手能够得到的地方都去摸一摸。 7但是这个也不是完全凭找东西就可以通关的。要靠线索去联想怎么回事。而且,如果没有用到的东西一定要把它带在身上,很多没用到的下一关就有可能用。工作人员会发给你传呼器,是给你提示用的,你们想不出来,就可以问问他。 选择加盟魔魔岛密室逃脱的5大理由 理由1:逼真代入感,如侦探小说一般,抽丝剥茧,环环相扣的逻辑,挑战你的IQ与思维。理由2:中型线索类密室最为考验玩家的逻辑推理能力,你不仅要具有当代福尔摩斯般敏锐的观察力和想象力,还要能勇敢的探索和决断。团队配合,将会是您胜出的关键! 理由3:体验过PC密室逃脱类游戏的玩家在体会找东西和解密快感的同时,还能体会到真实场景角色扮演的快乐。 理由4:各种梦幻场景,给玩家创造一个个惊喜。让玩家体会设计者的用心。 理由5:大型主题剧情机关解谜场景相结合,完美结合所有环节,更精致,更专业。

密室逃脱相关完整版

密室逃脱相关

Q1:别人做密室逃脱 从大二时,开始接触密室逃脱游戏,到今年寒假一天能玩上三、四次,“既然大家都喜欢,干嘛不自己也开一个呢?”黄皓月介绍,决定开一个“密室逃脱”店,很偶然,也很仓促。5个爱玩密室的女孩当即一拍即合,其中有两个女生已经工作,三个在校生中,黄皓月也即将本科毕业,她还“跨界”选修了金融专业的双学位。 三月份,考察江汉路所有密室逃脱店、玩遍40多个主题,敲定店址; 四月份,店面装修,自己设计主题、自己安装机关、自己制作装置……; “五一”,新店开张,首日客流量过百,不大的客厅里,挤满了前来“尝鲜”的同龄人。 开业的前一天,女孩们都在店里通宵加班,“创业比考研费精力多了,四月份的时候,天天熬夜。”黄皓月告诉记者,开店,比想象中的复杂、琐碎。对于“学霸”的称号,她觉得那是上高中以前的事了,从高二起,一直到现在,她更愿意把自己定义为“学霸班上的玩货”。 中考前,黄皓月因成绩优秀,提前保送到当地最好的学校——新洲一中“火箭班”。然而高一的第一次月考,成绩仅排在班上十几名,随后几次月考,更是滑落到二十几名。“我已经很努力了,学习成绩就是上不去。”家长的不理解,老师的不重视,让曾经的“神童公主”的骄傲光环瞬间破碎,从台上最闪亮的“明星”,跌落成台下普通观众,巨大的落差,让她一度“放松”自己,按照自己的意愿学习、生活,成为学霸班上的“另类”。 “其实二十几名已经很好了,至少可以考上武大华科,但那个时候没有人告诉我,我以为自己真的很差。”说到这里的时候,黄皓月的眼中有一丝伤感和落寞,虽然之后成绩时常徘徊在班上倒数几名,但高考总分依然超过一本分数线,考入湖北大学生物科学专业。 高中时期的那一段心路历程,对她触动很大,并且让“学霸班上的玩货”这一身份,一直保持到现在。今年,她们班上41名同学中,有三十几个考研,“没有同学相信我会考上,别人在学习的时候,我在玩。”她笑着说,作为前学生会副主席,如果她呆在寝室里安静的看书超过一周,所有人都会奇怪她没有不出去“活动”。 5月9日,小店开张后迎来的第一个双休日,女孩子们正在努力积累经验,希望在暑假旺季到来前,将自己打造成一个“全能女孩”。“我最大的优点,就是没有拖延症。”黄皓月觉得,“把今天做好”才是最重要的,至于最终结果是输是赢,只要自己努力过就好。她笑称,对未来没有明确的计划,如果读博的话,或许会考虑出国吧。 Q2采访5专家,2业主 业主1: 我是12年4月,5月的时候接触的密室逃脱,当时在武汉的EP逃逸层第一次接触到真人密室逃脱,当时整个武汉就两家,第一次体验了他们家的密室之后,我自己动了心思。6月我在创建了X-space真人密室逃脱。7月装修完开业,总共资金消耗6W不到,所有的谜题设计都是自己想的。那个时候也没什么房型啊,格局啊,什么的,一点纸片信息,一些文字内容,一点点的氛围,就用上房东家的家具,我们开业了。每天发发传单,做做问答,一个月的,收回了全部的成本还有不少的收益。当时也动了心思,想看看其他地方的密室逃脱是怎么做的,下一步,应该怎么发展。 永远要记住一点,你的视线永远要看着前方,比你能看到的位置更远的位置,然后收回来看脚下离目标有多远,选好节点,去执行。 我们的创业路径上很强调一个词,节点。后面也会不停的提到。

相关文档
最新文档