efecto en celulas

Upload: rbkamor

Post on 06-Jan-2016

7 views

Category:

Documents


2 download

DESCRIPTION

Efecto de los nanos en las celula

TRANSCRIPT

  • 1

    3/15/07 THE EFFECTS OF NANO CURRENT ON CELLULAR LIFE By Xanya Sofra-Weiss, Ph.D Microcurrent (1 microamp= 1 over a millionth of an ampere) and nano current (1 nano ampere = one over a billionth of an ampere) are known to influence biological processes at the cellular level by virtue of their similarity to physiologic actions, such as depolarization of cell membranes (Hodgkin, Huxley, 1952). Microcurrent has been reported to:

    - speed up wound healing by increasing cell proliferation (Goldman, Pollack, 1966).

    - Stimulate cells to proliferate but inhibit proliferation after a certain threshold (Ross, 1990; Blumenthal et al, 1997).

    - Increase cell proliferation, DNA and protein content (Yin et al, 2005) - Act as an antioxidant in chronic ulcers healing in human subjects (Bok

    et al, 2005) After a detailed analysis of the literature, certain articles were selected to enrich our understanding of micro electricity in order to form hypotheses for future research. Although micro electrical procedures have yet to be recognized as mainstream medical treatments, recent studies based on electronic advances reliably replicate results pointing towards the cability of micro electricity to resonate the wisdom of our inherent capacity for self regeneration and healing. Micro Current Effect on Cell Proliferation, DNA and Protein Content Yi-lo Lin et al (2005) used tissue cultures of tendon fibroblasts or tenocytes (the principal cellular components of tendon tissue) taken from 20 horses. Cells from these cultures were used for microcurrent stimulation (METS) experiments. The METS devise delivered a constant current, which means that the device automatically changed voltage as necessary to deliver the same current when peripheral resistance changed. The waveform consisted of a brief monophasic square pulse, duration 0.8 milliseconds. The pulse frequency was 150 Hz. Electrical current consisted of 0, 0.05, 0.1, 0.5, and 1.5ma or miliamps (1 milliamp = one over a thousand of an ampere). Application of microcurrent was via electrodes attached to paper strips soaked in the PBS solution that were placed in the culture wells. These investigators found that application of microcurrent had a stimulatory effect on cell proliferation which was significantly increased with repeated microcurrent applications (figure 1). Same results were observed for DNA content, except that application of current only once did not have a significant content compared with the control sample. However, repeated

  • 2

    microcurrent application significantly increased DNA content (Figure 2).

  • 3

    Figure 1 Effect of METS on proliferation of tenocytes in culture

    Figure 2 Effect of METS on DNA content of tenocytes in culture

  • 4

    Protein content significantly increased after one application of 0.5 and 1ma, and after two applications of 0.1, 0,5, and 1ma. However, application of microcurrent three times, significantly decreased protein content. Apoptosis rate did not alter after the first application. However after the third microcurrent application apoptosis rate significantly increased with increasing current intensity, so that the highest rate of apoptosis occurred at 1.5ma. The results of this study provides some evidence for the positive effects of microcurrent on cell proliferation, DNA and protein content, however, it raises questions as to the specifications and specific ranges of microcurrent necessary to produce optimum cell proliferation, DNA and protein content, while minimizing apoptosis (cell death). Cheng et al (1982) who studied the effects of miliamp and microcurrent on ATP generation protein synthesis and membrane transport showed that although low microcurrent stimulated physiologic activity of damaged cells and increased ATP up to 500%, ATP progressively decreased at miliamp ranges and dived down to 0 around 1.5ma. Therefore, the cell apoptosis observed in Yi-lo Lin et all study (2005) cited above, that appeared to be maximum with repeated applications of 1.5 ma, may well be the result of miliamp ranges depleting ATP. Additionally, the frequency of 150 Hz used in Yi-lo Lin study (2005) appears to contradict a number of experimental studies that report tissue healing within 1-4Hz band (Hefferman 1996). Santos et al (2004) showed epithelial regeneration, increased number of fibroblasts, collagen production and epidermis thickening at ultra low frequencies 0.5Hz. Lee at al (2005) used ultra low microcurrent ranging from 3 milliamps to 400 nano amperes at low frequencies that prevented the electrons from traveling in short bursts to heal chronic lesions in human subjects (see below under nano current as an antioxidant). In light of the above evidence, more research is necessary in testing cell proliferation, DNA and protein content and rate of apoptosis by using:

    1. Ultra low microcurrent ranging from 500 microamps down to 100 nano amperes.

    2. Ultra low frequencies, from 4.0 0.1 Hz that can prevent the electrons from traveling in short bursts.

    3. Constant current. Constant current is usually the result of using a constant current generator that adjusts the voltage when resistant changes. According to Ohms Law, Current is equal to Voltage over Resistance (I=V/R). In order to maintain I constant, the voltage needs to increase or decrease as resistance increases or decreases. For example if I=1 then V=R. If I=2 then 2V=R or V=1/2R and so on.

    There is an additional fourth component that may be a pertinent electrical specification component: the microcurrent waveform. Neher (Nobel Prize in Physiology and Medicine, 1991) observed square electrical signals emitted by the cell membrane. According to Mel C Siff (2000) microcurrent appears to operate more on the basis of resonant attunement of the stimulus to cellular

  • 5

    and subcellular processes and its effects depend on how efficiently the stimulation parameters match the electrical characteristic of the different cells. This resonant attunement may be impossible when the device merely produces a simple square waveform which is a sine waveform with a flat top that contains many higher frequencies, called harmonics. Harmonics can cause buzzing or other noise problems with some equipment. A clean complex square waveform that is composed out of several frequencies that stabilize it in the face of both electronic and biological resistances is necessary to produce the high definition signal that will tune into and resonate the cells biological processes. Nano current as an antioxidant Subatomic particle physics and the electron theory of electrical current comprise an explanation of how electrical nano currents (in the billionths of an ampere) can function as an antioxidant. An atom is made up of protons, neutrons and electrons. When molecules with weak bonds split, they can leave atoms with unpaired electrons, i.e. free radicals. Free radicals attack stable molecules to steal their electrons. When the attacked molecule loses an electron, it becomes a free radical itself, starting a chain reaction. Oxygen is attracted to a magnet, because oxygen has two unpaired electrons in its lowest energy state (Millikan, 1925). The existence of unpaired valence electrons in a stable molecule confers high chemical reactivity. Highly reactive molecules can oxidize molecules that were previously stable (oxidation refers to a loss of electrons) and turn them into free radicals. The food we eat is oxidized to produce high-energy electrons that are converted to stored energy. This energy is stored in high-energy phosphate bonds in the form of ATP (Adenosyne tri phosphate is a molecule of the structural unit of nucleotide chains forming nucleic acids as RNA and DNA). In the absence of oxygen, one molecule of glucose will yield four molecules of ATP. In the presence of oxygen we get 24 to 28 molecules of ATP from one molecule of glucose going through the Krebs cycle plus the four molecules from glycolysis. This entire oxidation process takes place in the mitochondria, making the system susceptible to the formation and effects of free radicals. A certain amount of oxidative function is necessary for proper health. For example, oxidative processes are used by the immune system to kill microorganisms. Overall, however, accumulated free radicals are one of the major contributing factors to aging and disease. Accumulated free radicals can even induce local injury to patients with poor immune systems, by reacting with lipids, proteins and nucleic acids, leading to cellular membrane damage. Antioxidants (eg. Vitamin C) are chemicals that have the ability to donate

  • 6

    electrons without becoming free radicals themselves. Nano amperes act as a powerful antioxidant by infusing a steady stream of electrons to the free radicals, ending the electron stealing chain reaction caused by free radicals. This was recently hypothesized in a study by Bok Y. Lee, MD, FACS, Alfred J. Koonin, M.B., Ch, B., Ph.D., FRCS, Keith Wendell, Ph.D., and John Hillard, RN, who used ultra low microcurrent (from 3 miliamp to 400 nano amperes) combined with low frequencies and an adjustable voltage to attain constant current, to treat 25 chronic skin ulcers present for an average of 18.5 months and not responding to standard conservative treatment in a hospital setting. Most lesions were in stages III and IV. Patients age varied from 20 to 85 years old. The lesions had the following chronic etiologies:

    - AIDS - Arterial Insufficiency - Cerebro-Vascular Accident - Chronic Obstructive Pulmonary Disease - Chronic Renal Failure - Congestive Cardiac Failure - Spinal Cord Injury - Traumatic Brain Injury - Venous Stasis

    Bok et al (2005) reported that 100% of the lesions treated with nano current showed response to the treatment. 100% healed in a maximum time of 7 weeks. Average time of healing was 48 hours of treatment over 16 days. Surgical debridement was unnecessary as the necrotic tissue appeared to disappear spontaneously. It has also shown that the lower the frequency the more permanent the effects of nano current. None of the healed lesions showed any sign of recurrence after a period ranging from six to eighteen months. (see results on figure 1) Many studies have shown that the rate of wound healing of an individual is directly proportional to their age. From this study it can be seen that treating chronic skin ulcers with ultra-low microcurrent (nano amperes) eliminates the age factor by equalizing the healing rate at all ages. The only limiting factor in healing time with this method seems to be the duration of the lesion. Overall Lee et als study (2005) indicates that a steady flow of electrons in a relatively low concentration appears to act exactly as one would expect from any antioxidant. The fact that the electrons are focused on a small region of the body may explain why healing changes appeared so rapidly. Lee et al (2005) concluded that the actual regeneration of the tissue coupled with the absence of the age factor in healing and the concomitant improvement noticed in the patients general condition all point to a highly potent antioxidant effect on the local tissues as well as generally.

  • 7

    Figure 3 -- Example of chronic lesion healing within an average of 16 days

  • 8

    What are the Microcurrent and Nano Current Effects on Cell Mitosis The Cell Cycle consists of several phases. In the first phase, (G1) the cell grows and becomes larger. After cytokinesis (the process where one cell splits off its daughter cell) the cells are quite small and low on ATP. They acquire ATP and increase in size during the G1 phase. After acquiring sufficient size and ATP, the cells enter the next phase (S) where they undergo DNA synthesis and a copy or each chromosome is formed. Since the formation of new DNA is an energy draining process, the cell undergoes a second growth and energy acquisition stage, the G2 phase. The energy acquired during G2 is used in cell division or mitosis. For all organisms it is essential that the different phases of the cell cycle are precisely coordinated. The phases must follow in the correct order, and one phase must be completed before the next phase can begin. Errors in this coordination may lead to chromosomes being lost, rearranged or distributed unequally between the two daughter cells. This type of chromosome alteration is often seen in cancer cells (Hartwell, Hunt and Nurse Nobel lecture 2001). Cheng (1982) reported a significant increase of ATP, up to 500%, following microcurrent stimulation. However, Chengs study did not explore:

    - the effects of increased ATP on cellular proliferation, - the effects of increased ATP on the proper coordination of cell division.

    The phase of the cell cycle may be important to the effects of microcurrent as cell cycle seems to influence the receptivity of the cell to extracellular signals. Generally, cells in the G0 / G1 stage undergo division when they receive signals that instruct them to enter the active phases of the cell cycle (Morgan et al, 2002). Deregulation of the cell cycle components has been shown to induce mitotic catastrophe and also may be involved in triggering programmed cell death of apoptosis (King et al, 2004). In conclusion, more research is necessary to test the hypothesis that microcurrent may have an effect on the coordination of cell division by virtue of increasing ATP. A even more interesting research study may be examining the effects of nano current resonance with biological signals instructing cells in the G0 / G1 stage to undergo division, by testing different complex waveforms (built with several frequencies) that vary in shape or composition. Can Nano Current have an effect on how Proteins Create Life? Cell biology research has revealed that although the cell nucleus reflects the cells reproductive system while the membrane represents its brain (Pray 2004, Silverman 2004, Lipton, 2005). Following enucleation (removing the nucleus to the cell) many cells can survive for up to two or more months,

  • 9

    demonstrating intelligence in their actively ingesting and metabolizing nutrients while performing all necessary physiological functions such as respiration, excretion, motility, etc. Enucleated cells die, not because they have lost their brain but because they have lost their reproductive capability. When you destroy the membrane, however, the cell dies, just as one would if ones brain was removed. Even if the membrane is left intact and you use digestive enzymes to destroy its receptor or effector proteins (Integral Membrane Proteins IMPs), the cell intelligent behavior ceases, and the cell becomes comatose. These findings have let scientists to the conclusion that the Integral Membrane Proteins (IMPs) are the fundamental units of cellular intelligence that control behaviors such as respiration, nourishment and waste (Lipton, 2005). IMPs are divided into receptor proteins that provide an awareness of environmental signals and effector (action) proteins. The receptor (awareness) - effector (action) proteins or IMPs act as a switch, translating environmental signals into cellular behavior. Studying how IMPs work has now become a field of its own called signal transduction. Signal transduction scientists focus on classifying hundreds of environmental signals and the activation of the cells behavior proteins. There are different kinds of effector proteins due to the variety of functions necessary for the smooth functioning of the cell. For example transport proteins include channel proteins that drive molecules and information from one side of the membrane barrier to the other. When the electrical charge of channel proteins is altered, the protein changes shape, allowing an open channel running through the proteins core that allows nutrients to go through the cell. In their inactive state, channel proteins are shaped like a tightly wound sphere, closed to the world outside the cell. A specific channel type sodium-potassium protein is called ATPase. This turns the cell into a constantly recharging biological battery. Another variety of effector proteins, the cytroskeletal proteins, regulates the shape and motility of the cells. A third variety called enzymes, breaks down or synthesizes molecules. When activated, all types of effector proteins provide signals that activate genes. IMPs and their byproducts provide signals that control the binding of the chromosomes regulatory proteins that form a sleeve around the DNA, thus controlling the reading of the genes. It takes about 100,000 different proteins to run our bodies. Each protein is a linear string of linked amino acid molecules, joined like a string of beads. The amino acids are linked by virtue of their electromagnetic positive and negative charges (like charges repel each other and opposite charges attract each other). The forces generated by the amino acids positive and negative charges bend and fold the flexible string of amino acids into what turns to be the most crucial elements of cellular life. The distribution of electromagnetic charge within a protein can be selectively altered by electromagnetic fields. Receptor (awareness) proteins antennas can read micro electrical signals. When these signals resonate with the receptors antennas, it will change the receptor proteins charge, causing it to change shape (Tsong 1989).

  • 10

    Analogues resonance processes may apply to effector (action) proteins. The mere fact that the life of the entire cellular system is based on electromagnetic charges, preludes another hypothesis yet to be tested by research. The hypothesis primarily revolves around the conformations (final shapes) of proteins and how nano current may be involved in the shape-shifting movements of proteins that propel cellular life. More specifically nano current may affect: (1) the proteins conformation changes that may generate movement to fulfill

    the cells physiological functions (eg. respiration or digestion); (2) the proteins cooperation in specific physiological functions known as

    pathways, including the energy-generating Krebs cycle; (3) the proteins proper folding, necessary for optimal functioning.

    Improperly folded proteins are marked for destruction by the cell, their backbone amino acids are disassembled and recycled in the synthesis of new proteins.

    Considering the importance of resonance between nano signal and a proteins antennas, a key aspect in testing the above hypotheses will be the specifications of the nano electricity used. As mentioned above the device used must be capable of the following specifications:

    1. Ultra low frequencies 2. Range of microcurrent from 500 micro amps (research reports an

    increase of ATP up to 500%) to 100 nano amperes 3. Constant current 4. A variety of complex waveforms that consist of a number of

    frequencies to ensure waveform accuracy and stability (eg. complex sine, square or rectangular waveforms.)

    An example of such a device is the Perfector, a machine primarily used in the field of cosmetics for non-invasive face lifts, scar healing and to reduce pigmentations, melasma, acne and rosacea. For more information on the Perfector go to www.arasysperfector.com and click on the science section.

  • 11

    REFERENCES: 1. Burnstock g, mc mahon sB, humphrey PPa, hamilton sg. - aTP (P2x) Receptors and Pain proceedings of the 9th World Congress on Pain. Progress in Pain Research and management 2000 vol.16, edited by divor m, Rowbotham mC, Wiesenfeld- hallin z, iasP Press, seattle 63-76. 2. dubyak gR, el-moatassim C. - signal transduction via P2-purinergic receptors for extracellular aTP and other nucleotides. am J Physiol 1993;265:C577-C606. 3. ding y, Cesare P, drew l, nikitaki d, Wood Jn. - aTP, P2x receptors and pain pathways. J auton nerv syst 2000; 81(1-3):289-294. 4. kennedy C. - The discovery and development of P2 receptor subtypes. J auton nerv syst 2000;81(13):158-163. 5. Communi d, govaerts C, Parmentier C, Boeynaems J-m. - Cloning of human purinergic P2y receptor coupled to phospholipase C and adeylyl cyclase. J Biol Chem 1997; 272:31969-31973. 6. kennedy C, Qi a, nicholas Ra, harden Tk. - differential coupling of human P2y11 receptor to phospholipase C and adeylyl cyclase. Br J Pharmacol 1999;126:22. 7. ostrom Rs, gregorian a, insel Pa. - Cellular release of and response to aTP as key determinants of the set-point of signal transduction pathways. J Biol Chem 2000;275(16):11735-11739. 8. di virgillo f. - dr. Jekyll/mr. hyde: the dual role of extracellular aTP> J auton nerve syst 2000;(1-3):59-63. 9. Bland-Ward Pa, humphrey PPa. - P2x receptors mediate aTP-induced primary nociceptive neurone activation. J auton nerv syst 2000;81(1-3):146-151. 10. hamilton sg, mcmahon sB. - aTP as a peripheral mediator of pain. J auton nerve syst 2000;81(1-3):187194. 11. souslova v, Cesare P, ding y, et al. - Warm-coding deficits and abherrant inflammatory pain in mice lacking P2x3 receptors. nature 2000;407:1015-1017. 12. seegers JC, engelbrecht Ca, van Papendorp dh. - activation of signal-tranduction mechanisms may underlie the therapeutic effects of an applied electric field. med hypoth 2001;accepted for publication. 13. ulett ga, han J, han s. - electroacupuncture: mechanisms and clinical application. Biol Psychiatry 1998;44:129-1382

  • 12

    14. van Papendorp dh, maritz C, dippenaar n. - Plasma levels of beta-endorphin, substrate P and leu encephalin in patients with chronic pain. accepted for publication oct/nov 2001. The medicine Journal. 15. allen BJ, menning Pm, Rogers sd, dhilardi J, mantyh PW, simone da. - Primary afferent fibres that contribute to increased substance P receptor internalization in the spinal cord after injury. J neurophysiol 1999;81(3):1379-1390. 16. nam Ts, Baik eJ, shin yu, Jeong y, Paik ks. - mechanism of transmission and modulation of renal pain in cats; effects of transcutaneous electrical nerve stimulation. yonsei med J 1995;36(2):187-201. 17. sluka ka, deacon m, stibal a, strissel s, Terpstra a. - spinal blockade of opioid receptors prevents the analgesia produced by Tens in arthritic rats. J Pharmacol exp Ther 1999;289(2):840-846. 18. ferguson dR, kennedy i, Burton TJ. - aTP is released from rabbit urinary bladder epithelial cells by hydrostatic pressure changes- a possible sensory mechanism. J Physiol 1997;505(2):503-511. 19. Cho mR, Thatte hs, silva mT, golan de. - Transmembrane calcium influx induced by ac electric fields. faseb J 1999;13:77-683. 20. mcCaig Cd, dover PJ. - Raised cyclic-amP and a small applied electric field influence differentiation, shape, and orientation of single myoblasts. dev Biol 1993;158(1):172-182. 21. lorich dg, Brighton CT, gupta, et al. - Biochemical pathway mediating the response of bone cells to capacitive coupling. Clin orthop 199;350:246-258. 22. Basset Cal. - Beneficial effects of electromagnetic fields. J Cell Biochem 1993;51:387-393. 23. leo kC, dostal Wf, Bossen dg, et al. - effect of transcutaneous electrical nerve stimulation characteristics on clinical pain. Physical Therapy 1986;6:200-205. 24. delitto a, strube mJ, shulman ad, et al. study of discomfort with electrical stimulation. Physical Therapy 1992; 72:410-424. 25. melzack R, Wall P. Pain mechanisms: a new theory. science 1965; 150:971. 26. Bauer W. electrical treatment of severe head and neck cancer pain. arch otolaryngol 1983; 109:382-383.

  • 13

    27. Cheng n. van hoff h, Bockx e, et al. The effect of electric currents on aTP generation protein synthesis, and membrane transport in rat skin. Clin, orthop 1982; 171:264-272. 28. kubler d, Pyerin W, kinzel v. Protein kinase activity and substrates at the surface on intact hela cells. J Biol Chem 1982; 257:322-329. 29. Pelleg a, Porters. The pharmacology of adenosine. Pharmacotherapy 1990; 10:157-174. 30. abbracchio mp, Burnstock g. Purinergic signalling: patho-physiological roles. Jpn J.pharmacol 1998; 78:113-145. 31. Burnstock g. Purinergic nerves. Pharm. Rev. 1972; 24:509-581. 32. greenberg s., di virgilio f, steinberg Th, silverstein sC. extracellular nucleotides mediate Ca2+ fluxes in J774 macrophages by two distict mechanisms. J Biol Chem. 1988; 263:10337-10343. 33. kanno s., oda n., abe m, saito s, hori k, handa y Tabaashi k and sato y. establisment of a simple and practical procedure applicable to therapeutic angiogenesis. Circulation 1999; 99(20):2682-2687. 34. fitzsimmons RJ, strong dd, mohan s, Baylink dJ. low-amplitude, low frequency electric field-stimulated bone cell proliferation may in part be mediated by increased igf-ii release. J Cell Physiol 1992; 150:84-89. 35. guigni Td, Braslau l, haigler hT. electric field-induced redistribution and post field relaxation of epidermal growth factor receptors on a431 cells. J Cell Biol 1987; 104:1291-1297. 36. goldman R and Pollack s. electric fields and proliferation in a chronic wound model. Bioelectromagnetics 1996; 17(6):450-457. 37. korensein R, somjen d, fischler h, Binderman i. Capacitative pulsed electric stimulation of bone cells. induction of cyclic-amP changes and dna synthesis. Biochim Biophys acta 1984; 803(4):302-307. 38. kirsch d, lerner f. innovations in pain management: a practical guide for clinicians. in: Weiner Rl (ed) electromedicine; deutsche Press 1990; 23:1-29. 39. Windsor Re, lester JP, herring sa. electrical stimulation in Clinical practice. Physician & sportsmedicine 1993; 21:85-93. 40. Reich Jd, Tarjan PP. electrical stimulation of skin. int J derm 1990; 29:395-400.

  • 14

    41. vodovnik l, karba R. Treatment of chronic wounds by means of electric and electromagnetic fields. a literature review. med Biol engineer Comput. 1992; 30:257-266. 42. stanish Wd et al. The use of electricity in ligament and tendon repair. Physician & sports medicine 1985; 13109-116. 43. Bailey, s. how microcurrent stimulation produces aTP - one mechanism. 2001 http://www.chiroweb. com/archives/17/18/index.html 44. katsugari T, Tokunaga T, ohba m, sato C, furukawa T. implication of aTP released from atrial, but not papillary, muscle segments of guinea pig by isoproterenol and forskolin. life sci 1993; 53:961-967. 45. lader as, xiao yf, oRiordan CR, Prat ag, Jackson gR Jr, Cantiello hf. camP activates an aTP-permeable pathway in neonatal rat cardiac myocytes. am J Physiol Cell Physiol 2000; 279(1):C173-187. 46. Roman Rm, Wang y, lidofsky sd, et al. hepatocellular aTP-binding cassette protein expression enhances aTP release and autocrine regulation of cell volume. J Biol Chem 1997; 272:21970-21976. 47. horibe e k. esttica clnica & cirrgica. in: horibe ek uso do cido tricloroactico. 1ed. Rio de Janeiro: Revinter; 2000. p 47-60. 48. Barnes TC. healing rate of human skin determined by measurement of the electrical potential of experiment abrasions. am J surg 1945;69:82 49. assimacopoulos d. Wound healing promotion by the use of negative electric current. am surg 1968; 4:423-31. 50. Cheng n, van hoff h, Bockx e. The effect of electric currents on aTP generation protein synthesis, and membrane transport in rat skin. Clin orthop 1982;171:264-72. 51. sasaki gh, Pang Cy. Pathophysiology and skin flaps raised on expanded pig skin. Plast Reconstr surg 1984; 4:59-69. 52. neter, J. applied linear statistical models. 4ed. Chicago: iRWin; 1996. 53. alvarez om, mertz Pm, smerbeck Rv. The healing of superficial wounds in stimulated by external electrical current. J invest dermatol 1983;81:144- 8. 54. american academy of medical accupuncture, 2001 What is acne? how does acne develop? 55. Carley PJ, Wainaipel sf. electrotherapy for acceleration of wound healing: low intensity direct current. arch Phys med Rehabil 1985;66:443-6.

  • 15

    56. Brown m, gogia PP. effects of high voltage stimulation on cutaneous wound healing in rabbits. Phys Ther 1987;67:662-7. 57. dunn mg. Wound healing using a collagen matrix: effect of dC electrical stimulation. J Biomed mater Res appl Biomat 1988;22:191-206. 58. kloth lC, feedar Ja. acceleration of wound healing with high voltage monophasic pulsed current. Phys Ther 1988; 68:503-8. mertz Pm, davis sC, Cazzaniga al, Cheng k, Reich Jd, eaglstein vm. electrical stimulation: acceleration of soft tissue repair by varying the polarity. Wounds 1993;5:153-9. 59. vodovnik l, karba R. Treatment of chronic wounds by means of electric and electromagnetic fields. Rev med Biol eng Comput 1992;30:257-66. 60. Electric microcurrent in the restoration of the skin undergone a trichloroacetic acid peeling in rats1 Microcorrente eltrica na restaurao da pele submetidos ao peeling de cido tricloroactico em ratos vilma natividade silva santos2, lydia masako ferreira3, edith kawano horibe4, ivone da silva duarte4, 1994. study developed in the Post-graduation Program of Recovering Plastic surgery of the federal university of so Paulo (unifesP) Paulista school of medicine (ePm). so Paulo Brazil. 61. Correspondence: vilma natividade silva santos departamento de Cirurgia Plstica Rua napoleo de Barros, 715 04024-002 so Paulo sP Brazil Tel: (55 11) 5576-4065 / 5576-4118 [email protected] Received: may 4, 2004 62. 2004 sTCC foundation Press written by dawn a. Tamarkin, Ph.d. 63. Charles J. Currey, mha, Pa-C university of florida Pa Program, summer, 2005 64. Science News, March 30, 1991, p 207 65. longevity, december, 1992, p 14 66. houston Post, march, 20 1991 67. isopathic/homeopathic materia medica,by Werthmann and schneider 1999 Published by semmelweisverlag (germany) 68. Beck Protocol, august 2000, available by contacting the eagle foundation (Rose stevens) (204)-2543996 69. The Cancer Cure that Worked, Barry lynes, 1987, isBn 0-919951-30-9 70. nexus, september/october, 2001, p 17

  • 16

    71. sinitsyn, Razvozva (Russian): effects of electrical microcurrents on Regeneration Processes in skin Wounds ortop Travmatol Protez, feb. 1986 72. sinitsyn, Razvozova, (Russian): stimulation of the Regeneration of skin Wounds by microcurrents vopr Juroortol fizioter lech fiz kult, nov.-dec. 1985 73. guyton aC, hall Je. Textbook of medical Physiology. W.B. saunders Company. 74. kroschwitz Ji, Winokur m. Chemistry - general, organic, Biological. mcgraw-hill, inc. 75. giancoli dC. Physics: Principles with applications. Prentice hall. 76.Carley and Wainapel. electrotherapy for acceleration of wound healing: low intensity direct current. archives of Physical medicine and Rehabilitation July 1985; vol. 66. 77. Becker Ro. Cross Currents: The Perils of electropollution, the Promise of electromedicine. g.P. Putnams Sons. 78. Becker.Ro: The Body electric. new york 79. William morrow and Co. inc. 1983 80. nordenstrom.B: Biologically Closed electrical Circuits, nordic medical Publications. uppsala. 1983. 81. Wallace. la. mens Therapy: Clinical Perspectives vol. 1. Cleveland: privately published. 1990. distribution. 82. Bourgingon g, f.a.s.e. b.5 398-4021987 electrical stimulation of protein and d.n.a. synthesis in human fibroblasts. 83. almeida-Porada g, Porada Cd, Chamberlain J, Torabi a, zanjani ed (2004) formation of human hepatocytes by human hematopoietic stem cells in sheep. Blood 104: 25822590 84. amit m, shariki C, margulets v, itskovitz-eldor J (2004) feeder layer- and serum-free culture of human embryonic stem cells. Biol Reprod 70: 837845 85. Bach fh, albertini RJ, Joo P, anderson Jl, Bortin mm (1968) Bone-marrow transplantation in a patient with the Wiskottaldrich syndrome. lancet 2: 13641366 86. Bjerknes m, Cheng h (1999) Clonal analysis of mouse intestinal epithelial progenitors. gastroenterology 116: 714

  • 17

    87. Cerami a (1985) hypothesis: glucose as a mediator of aging. J am geriatr soc 33: 626634 88. Chen J (2004) senescence and functional failure in hematopoietic stem cells. exp hematol 32: 1025 1032 89. Clark WR (1999) a means to an end: The Biological Basis of aging and death. new york, ny, usa: oxford university Press 90. davenport RJ (2004) Regenerating regeneration. sci aging knowl environ 2004 91. finkel T, holbrook nJ (2000) oxidants, oxidative stress and the biology of ageing. nature 408: 239247 92. gage fh (2000) mammalian neural stem cells. science 287: 14331438 93. hayflick l (1994) how and Why We age. new york, ny, usa: Ballantine Books 94. hayflick l, moorhead Ps (1961) The serial cultivation of human diploid cell strains. exp Cell Res 25: 585621 95. ho ad, Punzel m (2003) hematopoietic stem cells: can old cells learn new tricks? J leukoc Biol 73: 547555 96. korbling m, dorken B, ho ad, Pezzutto a, hunstein W, fliedner Tm (1986) autologous transplantation of blood-derived hemopoietic stem cells after myeloablative therapy in a patient with Burkitts lymphoma. Blood 67: 529532 97. mahlknecht u, hoelzer d (2000) histone acetylation modifiers in the pathogenesis of malignant disease. mol med 6: 623644 98. maximow a (1909) der lymphozyt als gemeinsame stammzelle der verschiedenen Blutelemente in der embryonalen entwicklung und im postfetalen leber der sugetiere. folia haematol (leipzig) 8: 125141 99. mcCormick a, Campisi J (1991) Cellular aging and senescence. Curr opin Cell Biol 3: 230234 100. morshead Cm, Benveniste P, iscove nn, van der kooy d (2002) hematopoietic competence is a rare property of neural stem cells that may depend on genetic and epigenetic alterations. nat med 8: 268273 101. siminovitch l, mcCulloch ea, Till Je (1963) The distribution of colony-forming cells among spleen colonies. J Cell Physiol 62: 327336

  • 18

    102. Terada n et al (2002) Bone marrow cells adopt the phenotype of other cells by spontaneous cell fusion. nature 416: 542545 103. Thomas ed, flournoy n, Buckner Cd, Clift Ra, fefer a, neimen Pe, storb R (1977) Cure of leukemia by marrow transplantation. leukemia Res 1: 6770 104. Thomson Ja et al (1998) embryonic stem cell lines derived from human blastocysts. science 282: 11451147 105. Wagers aJ, Christensen Jl, Weissman il (2002) Cell fate determination from stem cells. gene Ther 9: 606612 106. Weissman il (2000) Translating stem and progenitor cell biology to the clinic: barriers and opportunities. science 287: 14421446 107. ying Ql, nichols J, evans eP, smith ag (2002) Changing potency by spontaneous fusion. nature 416: 545548 108. zanjani ed et al (1992) engraftment and long-term expression of human fetal hemopoietic stem cells in sheep following transplantation in utero. J Clin invest 89: 11781188 109. almers, W. & neher, e. (1985) feBs letters 192, 13-18. 110. almers, W. & neher, e. (1987)J. Physiol. 386, 205-217. 111. anderson, C.R. & stevens, C.f. (1973) J. Physiol. 235, 655-691. 112. augustine, g.J. & neher, e. 1992, J. Physiol. 450, 247-271. 113. augustine, g. J., Charlton, m.P. & smith, s.J. (1987) ann. Rev. neurosci. 10,633 114. Bean, R.C., shepherd, W.C., Chan, h., & eichler, J.T. (1969) J. gen. Physiol. 53, 741-757. 115. Berridge, m.J. & irvine, R.f. (1989) nature 341, 197-205. 116. Cockcroft, s. & gomperts, B.d. (1985) nature 314, 534-536. 117. Conti, f. & neher, e. (1980) nature 285, 140-143. 118. di virgilio, f., lew, d.P. & Pozzan, T. (1984) nature 310, 691-693. 119. fenwick, e.m., marty, a. & neher, e. (1981) J. Physiol. 319, l00P-101P 120. fenwick, e.m., marty, a. & neher, e. (1982a) J. Physiol. 331, 577-597.

  • 19

    121. fenwick, e.m., marty, a. & neher, e. (1982b) J. Physiol. 331, 599-635. 122. fernandez, J.m., neher, e. & gomperts, B.d. (1984) nature 312, 453-455. 123. fesenko, e.e., kolesnikov, s.s. & lyubarsky, a.l. (1985) 124. nature 313, 310-313. fishman, h.m. (1973) Proc. natl. acad. sci. usa 70, 876-879. frank, k. & Tauc, l. (1963) in: The Cellular function of membrane Transport (ed. J. 125. hoffman), Prentice hall, englewood Cliffs, new Jersey/usa gillespie, T.J. (1979) Proc. R. soc. london ser. B 206, 293-306. 126. gomperts, B.d. Barrowman, m.m. & Cockcroft, s. (1986) fed. Proc. 45, 21562161. hamill, o.P. (1983) in: single Channel Recording (eds. B. sakmann and e. neher) 127. Plenum Press, new york, pp 451-471. 128. hamill, o.P. & sakmann, B. (1981) J. Physiol. 312, 41P-42P. 129. J Periodontol. 1976 Jun;47(6):311-9. links Electrical stimulation of osteogenesis in pathological osseous defects. hamill, o.P., marty, a., neher, e., sakmann, B., & sigworth, f.J. (1981) Pfliigers 130. arch. 391, 85-100. haslam, R.J. & davidson, m.l. (1984) feBs letters 174, 90-95. 131. heiman, a.s. & Crews, f.T. (1985) J. immunol. 134, 548-554). hescheler, J., kameyama, m. & Trautwein, W. (1986) Pfliigers arch. 407, 182-189. 132. hille, B. (1970) Prog. Biophys. mol. Biol. 21, l-32. hille, B. (1989) Quart. J. exp. Physiol. 74, 785-804. 133. hladky, s.B. & haydon, d.a. (1970) nature 225, 451-453. 134. hodgkin, a.l. & huxley, a.f. (1952) J. Physiol. 117, 500-544. 135. horn, R. & marty, a. (1988) J. gen. Physiol. 92, 154-159. horn, R. & Patlak, J. (1980) Proc. natl. acad. sci. usa 77, 6930-6934. 136. Jaffe, l.a., hagiwara, s. & kado, R.T. (1978) dev. Biol. 67, 243-248. 137. sir Bernard katz (1966) nerv, muscle and synapse, mcgraw-hill Book Company, new york

  • 20

    138. kameyama, m., hescheler, J., hofmann, f., & Trautwein, W. (1986) Pfliigers arch. 407, 123-l 28. 139. katz, B. & miledi, R. (1972) J. Physiol. 224, 665-700. 140. kostyuk, P.g. (1980) neuroscience 5, 945-959. 141. Chi 2003 (university of Washinghton) 142. Chi 1999 (university of Washinghton) 143. Pflugers arch. 1991 may;418(4):423-9. gating currents of inactivating and non-inactivating potassium channels expressed in xenopus oocytes. stuhmer W, Conti f, stocker m, Pongs o, heinemann sh, Baldissera f, Campadelli P. how motoneurones control development of muscle tension. nature. 1977 Jul 14;268(5616):146147. [Pubmed] 144. Baldissera f, Campadelli Paola, Piccinelli l. neural encoding of input transients investigated by intracellular injection of ramp currents in cat -motoneurones. J Physiol. 1982;328:7386. [Pubmed] 145. Baldissera f, Campadelli P, Piccinelli l. The dynamic response of cat alpha-motoneurones investigated by intracellular injection of sinusoidal currents. exp Brain Res. 1984;54(2):275282. [Pubmed] Baldissera f, Parmiggiani f. Relevance of motoneuronal firing adaptation to tension development in the motor unit. Brain Res. 1975 Jun 27;91(2):315320. [Pubmed] 146. Burke Re, dum RP, fleshman JW, glenn ll, lev-Tov a, odonovan mJ, Pinter mJ. a hRP study of the relation between cell size and motor unit type in cat ankle extensor motoneurons. J Comp neurol. 1982 Jul 20;209(1):1728. [Pubmed] 147. Burke Re, levine dn, Tsairis P, zajac fe iii. Physiological types and histochemical profiles in motor units of the cat gastrocnemius. J Physiol. 1973 nov;234(3):723748.3. [Pubmed] 148. Cooper, sybil. eccles JC. The isometric responses of mammalian muscles. J Physiol. 1930 Jun 27;69(4):377385. [Pubmed] 149. eccles JC, eccles Rosamond m, iggo a, ito m. distribution of recurrent inhibition among motoneurones. J Physiol. 1961 dec;159(3):479499. [Pubmed]

  • 21

    150. fleshman JW, munson JB, sypert gW, friedman Wa. Rheobase, input resistance, and motor-unit type in medial gastrocnemius motoneurons in the cat. J neurophysiol. 1981 dec;46(6):13261338. [Pubmed] 151. grimby l, hannerz J. firing rate and recruitment order of toe extensor motor units in different modes of voluntary conraction. J Physiol. 1977 Jan;264(3):865879. [Pubmed] 152. gustafsson B, Pinter mJ. Relations among passive electrical properties of lumbar alpha-motoneurones of the cat. J Physiol. 1984 nov;356:401431. [Pubmed] 153. gustafsson B, Pinter mJ. an investigation of threshold properties among cat spinal alpha-motoneurones. J Physiol. 1984 dec;357:453483. [Pubmed] 154. harrison PJ. The relationship between the distribution of motor unit mechanical properties and the forces due to recruitment and to rate coding for the generation of muscle force. Brain Res. 1983 apr 4;264(2):311315. [Pubmed] 155. henneman e, somJen g, CaRPenTeR do. funCTional signifiCanCe of Cell size in sPinal moToneuRons. J neurophysiol. 1965 may;28:560580. [Pubmed] 156. henneman e, somjen g, Carpenter do. excitability and inhibitability of motoneurons of different sizes. J neurophysiol. 1965 may;28(3):599620. [Pubmed] 157. kernell d. input resistance, electrical excitability, and size of ventral horn cells in cat spinal cord. science. 1966 Jun 17;152(729):16371640. [Pubmed] 158. kernell d. Rhythmic properties of motoneurones innervating muscle fibres of different speed in m. gastrocnemius medialis of the cat. Brain Res. 1979 Jan 5;160(1):159162. [Pubmed] 159. kernell d. functional properties of spinal motoneurons and gradation of muscle force. adv neurol. 1983;39:213226. [Pubmed] 160. kernell d, monster aW. Threshold current for repetitive impulse firing in motoneurones innervating muscle fibres of different fatigue sensitivity in the cat. Brain Res. 1981 dec 14;229(1):193196. [Pubmed] 161. kernell d, zwaagstra B. input conductance axonal conduction velocity and cell size among hindlimb motoneurones of the cat. Brain Res. 1981 Jan 12;204(2):311326. [Pubmed]

  • 22

    162. kuno m. excitability following antidromic activation in spinal motoneurones supplying red muscles. J Physiol. 1959 dec;149(2):374393. [Pubmed] 163. milner-Brown hs, stein RB, yemm R. The orderly recruitment of human motor units during voluntary isometric contractions. J Physiol. 1973 apr;230(2):359370. [Pubmed] 164. Partridge ld. signal-handling characteristics of load-moving skeletal muscle. am J Physiol. 1966 may;210(5):11781191. [Pubmed] 165. Karen L. King, John A. Cidlowski Cell Cycle and Apoptosis: Common Pathways to Life and Death, Journal of Cellular Biochemistry, Volume 58, Issue 2 , Pages 175 180; 2004 166. Hartwell, Hunt and Nurse: Key Regulators of the Cell Cycle, Nobel Prize in Physiology and Medicine; 2001 167. Lipton B, H.: The Biology of Belief, 2005 168. Lipton B, H et al: Microvessel Endothelial Cell Transdifferentiation: Phenotypic Characterization Differentiation 46: 117-133. 169. Lipton B, H et al: :histamine-Modulated transdifferentiation of Dermal Microvascular Endothelial Cells. Experimental Cell Research 199: 279-291. Specific Microcurrent References 1. Aaron RK, Ciombor, D & Jolly G Stimulation of endochondral ossification by low-energy pulsing electromagnetic fields. J. Bone Mineral Res. 4:227-233; 1989. 2. Adey, RW. ELF magnetic fields and promotion of cancer: experimental studies In: B. Norden & C. Ramel (eds.) Interaction Mechanisms of low-level Electromagnetic Fields in Living Systems 1992: 23-46. Oxford University Press, Oxford. 3. Adey, RW. Electromagnetics in Biology and Medicine In: H. Matsumoto (ed) Modern Radio Science 1993: 177-245. Oxford University Press, Oxford. 4. Adey, RW. Signal functions of brain electrical rhythms and their modulation by external electromagnetic fields. In: E. Basar and T. Bullock (eds) Induced Rhythms of the Brain Birkauser, Boston, 1991: 323-351. 5. Adey, RW. Some fundamental aspects of biological effects of extremely low frequency (ELF). In: Grandolfo, M; Michaelson, S (eds.) Biological effects

  • 23

    and dosimetry of Ionizing Electromagnetic Fields. New York: Plenum Publishing; 1983:561-580. 6. Anderson, JC. & Eriksson, C. Piezoelectric properties of dry and wet bone. Nature 227:491--492; 1970. 7. Auerbach, GD.; Marx, SJ & Spiegel, AM. Parathyroid hormone, calcitonin and the calciferols. In: Wilson, JD & Foster, WD. (eds.) Williams' Textbook of Endocrinology. 7th ed. New York: Saunders; 1985:1137-1217. 8. Barnothy MF (ed) Biological Effects of Magnetic Fields Plenum Press 1969 9. Bassett CA Pulsing electromagnetic fields: A new approach to surgical problems. In: Buchwals H, Varco RL. Metabolic Surgery New York: Gune & Stratonn, 1982b:255-366. 10. Bassett CA Pulsing electromagnetic fields: A new method to modify cell behavior in calcified and noncalcified tissues. Calsif Tiss Int 1982; 34:1-8. 11. Bassett CA. Biologic significance of piezoelectricity. Calc Tiss Res 1968;1:252-72. 12. Bassett CA & Becker RO. Generation of electric potentials by bone in response to mechanical stress. Science 1962;137:1063-4 13. Bassett CA, Pawluck R & Becker RO. Effects of electric current on bone in vivo. Nature 1964;204:652-54. 14. Bassett CAL, Pawluk RJ, Pilla A. Acceleration of fracture repair by electromagnetic field. A surgically non-invasive method. Ann NY Acad Sci 1974;238:242-62 15. Bassett, CA. Biomedical implications of pulsing electromagnetic fields. Surg. Rounds 1983:22-31; 1983. 16. Bassett, CA Pulsing electromagnetic fields: A new method to modify cell behavior in calcified and noncalcified tissues. Calc. Tiss. Res. 34:1-8; 1982. 17. Bassett, CA. & Becker, RO. Generation of electric potentials in bone in response to mechanical stress. Science 137:1063-1064; 1962. 18. Bassett, CA & Hermann, I. The effect of electrostatic fields on macromolecular synthesis by fibroblasts in vitro. J. Cell Biol. 39:9a; 1968. 19. Bassett, CA.; Mitchell, S & Gaston, S. R. Pulsing electromagnetic field treatment in ununited fractures and failed arthrodeses. JAMA 247:623-628; 1982.

  • 24

    20. Bassett, CA; Pawluk, R.; Becker, RO. Effect of electric currents on bone in vivo. Nature 204:652-654; 1964. 21. Bassett, CA; Pawluk, R.J.; Pilla, AA. Acceleration of fracture repair by electromagnetic fields. A surgically non-invasive method. Ann. N Y Acad. Sci. 238:242-262; 1974. 22. Bassett, CA; Pilla, AA.; Pawluk, RJ. A non-operative salvage of surgically-resistant pseudarthroses and non-unions by pulsing electromagnetic fields. Clin. Orthop & Rel. Res. 124: 128-143; 1977. 23. Battocletti JH Electromagnetism, Man and the Environment Paul Elek, London 1976 24. Becker RO The basic biological data transmission and control system influenced by electrical forces Ann. N Y Acad. Sci. 1974, 238: 236-241 25. Becker, RO. The significance of bioelectric potentials. Bioelectrochem. Bioenerget. 1:187-199; 1978. 26. Becker, RO Cross Currents: The Promise of Electromedicine and the Perils of Electropollution New York: Putnam. 1990 27. Beeson DC, Johnston LE Jr, Wisotzky J. Effect of constant currents on orthodontic tooth movement in the cat. J Dent Res 1975;54:251-4. 28. Berridge, M. The molecular basis of communication within the cell. Sci. Am. 253:142-150; 1985. 29. Bjork A. Prediction of mandibular growth rotation. Am J Orthod 1969; 55:585-99. 30. Bjork A. Variations in the growth pattern of the human mandible: longitudinal radiographic study by the implant method. J Dent Res 1963;42:400-411. 31. Black J. Tissue response to exogenous electromagnetic signals. Orthop Clin N Amer 1984;15:15-31. 32. Borgens, RB. Endogenous ionic currents traverse intact and damaged bone. Science 225:478--482; 1984. 33. Bourguignon, Gerard J.; Wenche, J & Bourguigon Lilly W. Electric stimulation of fibroblasts causes an increase in calcium influx and the exposure of additional insulin receptors. J of Cellular Physiol 1989; 140:379-385.

  • 25

    34. Brighton CT, Black J & Pollack SR. Electrical properties of Bone and Cartilage: Experimental Effects and Clinical Applications. Grune & Stratton Inc. New York NY 1979. 35. Brighton CT, Heppenstall RB. Oxygen tension in bones of the epiphyseal plate, the metaphysis, and diaphysis. An in vitro and in vivo study in rats and rabbits. J Bone Joint Surg 1971; 53A:718-729. 36. Brighton CT, Hunt RM. Ultrastructure of electrically induced osteogenesis in the rabbit medullary canal. J Orthop Res 1986;4:27-36. 37. Brighton CT, Shaman P, Heppenstall R, Esterhai J, Pollack S, Friedenberg Z. Tibial nonunion treated with direct current, capacitive coupling, or bone graft. Clin Orthop 1995;321:223-34. 38. Brighton CT. Bioelectric effects on bone and cartilage. Clin Orthop & Rel Res 1977;124:2-4. 39. Brighton, CT. Bone reaction to varying amounts of direct current. Surg. Gynecol. Obstet. 131:894; 1970. 40. Brighton, CT (ed). Electrical properties of Bone and Cartilage. New York: Plenum Press; 1979:519-545. 41. Brighton, CT.; Black, J.; Friedenberg, Z.; Esterhai, J; Day, L; Cormoily, J. A multicenter study of the treatment of non-union fractures with constant direct current. J. Bone & Joint Surg. 63A:2-12; 1981. 42. Brighton, CT & Friedenberg, ZB Electrical stimulation and oxygen tension. Ann. N Y Acad. Sci. 238:314-320; 1974. 43. Brighton, CT; Friedenberg, ZB & Black, J. Evaluation of the use of constant direct current in the treatment of non-union. In: Brighton, CT (ed.) Electrical properties of Bone and Cartilage. New York: Plenum Press; 1979:519-545. 44. Brighton, CT.; Unger, A.; Starebough, J. In vitro growth of bovine articular cartilage chondrocytes in various capacitatively coupled electrical fields. J. Orthop. Res. 2:15-22; 1984. 45. Byl N, McKenzie A, West J, Whitney J, Hunt T, Holp H & Scheuenstuhl H. Pulsed microamperage stimulation: a controlled study of healing of surgically induced wounds in Yucatan pigs. Phys Ther 1994;74:201-13. 46. Cain, CD & Luben, R. Pulsed EMF effects on PTH stimulated cAMP accumulation and bone resorption in mouse calvariae. In: Anderson, L; Kelman, B; Weigel, R (eds) Interaction of Biological systems with ELF.

  • 26

    Richland, WA: Battelle Laboratories Press; Conference Publication No. 24; 1987, 269-278. 47. Cain, CD. Pulsed Electromagnetic Field modifications on Bone Metabolism in vitro: Influences on cyclic AMP ornithine decarboxylase and Bone Resorption. Riverside University of California, Dept of Biochemistry; Ph.D. dissertation. 1986 48. Cain, CD.; Adey, WR & Luben, RA. Evidence that. puIsed electromagnetic fields inhibit coupling of adenylate cyel by parathyroid hormone in bone cells. J. Bone Min. Res.I 437-441; 1987. 49. Cain, CD.; Luben, R. A. Pulsed EMF effects on PTH stimulated cAMP accumulation and bone resorption in mouse calvariae. In: Anderson, L; Kelman, B; Weigel, R (eds) Interaction of Biological Systems with ELF. Richland, WA: Battelle Laboratories Press; Conference Publication No. 24; 1987, 269-278. 50. Canalis, E. Regulation of bone remodeling. In: M. J. Fayus (ed) Primer on metabolic bone diseases. Kelseyville, CA: Amer Society for Bone & Mineral Research; 1990:23-25. 51. Carley & Wainapel. Electrotherapy for acceleration of wound healing: low intensity direct current. Arch of Physical Med & Rehab July 1985; vol. 66. 52. Chakkalakal DA, Lipello L, Shindell RL, Connoly JF. Electrophysiology of direct current stimulation of fracture healing in canine radius. IEEE Trans Biomed Eng 1990;37,11:1048-58. 53. Cheng, N, The effect of Electric Currents on ATP Generation, Protein Synthesis, And Membrane Transport in Rat Skin. Clin. Orthopedics & Rel Res 1982; 171: 264-272 54. Cheng, N., The effects of electric currents on ATP Generation, Protein Synthesis, and membrane transport in rat skin. Orth Surg. 1982 55. Childers KR. The Effects of Direct Electric Current on Condylar and Mandibular Growth in the Rabbit. Masters Thesis, University of Tennessee, 1990. 56. Clinical Applications of Electric Current remain largely unexploredJ Am Med Assoc 1974, 227: 129-130 57. Cochran, GV; Pawluk, RJ.; Bassett, CA. Electrum chanical characteristics of bone under physiologic moist conditions. Clin. Orthop & Rel. Res. 58:250-269; 1968.

  • 27

    58. Colacicco, G.; Pilla, AA. Chemical, physical and biological correlations in the Ca-uptake by embryonal chick tibia in vitro. Biochem. & Bioenerget. 10:119-131; 1983. 59. Compere, CL. Electromagnetic fields and bones. JAMA 247: 669; 1982. 60. Cone, CD. Unified theory on the basic mechanism of normal mitotic control and oncogenesis. J. Theor. Biol. 30:151-181; 1971. 61. Cook, I & Bassett, CA Effects of tissue type and orientation of electromagnetically induced voltages. J. Bone Joint Surg. 7:361-366; 1983. 62. Creekmoore TD & Radney L Frankel appliance therapy: orthopedic or orthodontic? Am J Orthod 1983;83:89-108. 63. Czech, M. Signal transmission by the insulin-like growth factors. Cell 59:235-238; 1989. 64. Darnell, J.; Lodish, H & Baltimore, D. Cell-to-cell Signaling. Hormones and Receptors. Molecular Cell Biology. 2nd ed. New York: W. H. Freeman; 1990:709-763. 65. Davidovitch Z, Finkelson M, Steigman S, Shanfield J, Montgomery P & Korostoff E. Electric currents, bone remodeling, and orthodontic tooth movement. The effect of electric currents on periodontal cyclic nucleotides. AM J Orthod 1980a;77:14-32. 66. Davitovitch Z, Finkelson M, Steigman S, Shanfield J, Montgomery P & Korostaff E. Electric currents, bone remodeling, and orthodontic tooth movement. II Increase in the rate of tooth movement and periodontal cyclic nucleotide by combined force and electric currents. AM J Orthod 1980b;77:33-47. 67. Davitovitch Z & Shanfield JL. Cyclic AMP levels in alveolar bone of orthodontically treated cats. Arch Oral Biol 1975;20:567-74. 68. De Loecker W & Stas. ML Effect or cortisol treatment on free amino acid levels in rats. J. Endocrinol. 59:57. 1973. 69. Dealler, S. F. Electrical phenomena associated with bones and fractures and the therapeutic use of electricity in fracture healing. J. Meal. Engin. Technol. 5:73-79; 1981. 70. Dietrich, J. W.; Canalis, E. M.; Maina, D. M. & Raisz, L. Hormonal control of bone collagen synthesis in vitro: Effects of parathyroid hormone and calcitonin. Endocrinology 98:943-949; 1976.

  • 28

    71. DoMman, HG.; Caron, M. G.; Lefkowitz, R. J. A family of receptors coupled to guanine nucleotide regulatory proteins. Biochem. 26:2657-2664; 1987. 72. Electric Current as a Bone Healer Med World News 1975, 16(2): 84 73. Electronic Medical Digest: San Francisco: Electronic Medical Foundation; 1944-1955 74. Epstein, S. Bone-derived proteins. Trends Endocr. Metab. 1:9-13; 1990. 75. Eriksson. C. Streaming potentials and other water-dependent effects in mlneralized tissues. Ann. N.Y. Acad. Sci. 238:321, 1974. 76. Eriksson. C Electrical properties of bone. In Bourne GH (ed.): Biochemistry and Physiology of Bone. vol. 4. New York. Academic Press, 1976, pp. 329-384. 77. Farndale RW, Maroudas A & Marsland T. Effects of low-amplitude pulsed magnetic fields on cellular ion transport. Bioelectromagnetics 1987;8,2:119-34. 78. Farndale RW & Murray JC. Pulsed electromagnetic fields promote collagen production in bone marrow fibroblasts via athermal mechanisms. Calcif Tissue Int 1985;37:178-82. 79. Ferrier J, Ross SM, Kanehisa J, Aubin JE. Osteoclasts and osteoblasts migrate in opposite directions in response to a contact electrical field. J Cell Physiol 1986;129:283-8. 80. Firzsimmons RJ, Strong D, Mohan S & Baylink D. Low-amplitude, low-frequency electric field-stimulated bone cell proliferation may in part be mediated by increased IGF-II release. J Cell Physiol 1992;150,1:84-9. 81. Fitton-Jackson, S & Bassett, C. A. The response of skeletal tissues to pulsed magnetic fields. In: Richards, R. J.; Rajah K. T. (eds) Use of Tissue Culture in Medical Research. Oxford Pergamon Press; 1980:21-46. 82. Florey, W & Neuhaus, O Induced transport of amino acids in rat liver after whole body irradiation. Radial. Res. 68:138, 1976. 83. Friedenberg ZB, Andrews ET, Smolenski BI, Pearl BW, Brighton CT. Bone reaction to varying amounts of direct current. Surg Gynec Obstet 1970;131:894-9. 84. Friedenberg ZB, Brighton CT. Bioelectric potentials in bone. J Bone Joint Surg 1966;48-A:915-23.

  • 29

    85. Friedenberg ZB, Brighton CT. Electric fracture healing. Ann NY Acad Sci 1974;238:564-74. 86. Friedenberg ZB, Dyer RH, Brighton CT. Electro-osteograms of long bones of immature rabbits. J Dent Res 1971a;50:635-9 87. Friedenberg ZB, Harlow MC, Brighton CT. Healing on nonunion of the medial malleolus by means of direct current: a case report. J Trauma 1971b; 11:883-5 88. Friedenberg ZB & Kohanim M. Effect of direct current on bone. Surg Gynec Obstet 1968;127:97. 89. Friedenberg ZB, Roberts PG Jr., Dedizian W, Brighton CT. Stimulation of fracture healing by direct current in the rabbit fibula. J Bone Joint Surg 1971c; 53A:1400-8. 90. Friedenberg, ZB & Brighton, CT. Bioelectric potentials in bone. J. Bone Joint Surg. 48A:915-923; 1966. 91. Friedenberg, ZB & Brighton, CT. Bioelectricity and fracture healing. Plast. Reconst. Surg. 68:435-443; 1981. 92. Friedenberg, ZB.; Harlow, MC & Brighton, C Healing of non-union of the medial malleolus by means of direct current: A case report. J. Trauma 11:883-885; 1971a. 93. Friedenberg, ZB.; Harlow, MC.; Heppenstali, R. B.; Brighton, C. T. The cellular origin of bioelectric potentials in bone. Calc. Tiss. Res. 13:53; 1973. 94. Friedenberg, ZB.; Roberts, P. G.; Didizian, N.H.; Brighton, C. T. Stimulation of fracture healing by direct current in the rabbit fibula. J. Bone Joint Surg. 53A: 1400-1408; 1971b. 95. Friedenberg, ZB.; Zemsky, L. M.; Pollis, R. P.; Brighton, C. T. The response of non-traumatized bone to direct current. J. Bone Joint Surg. 56A: 1023-1040; 1974. 96. Fukada E, Lang S, Mascarenhas S, Pilla A, Shamos M. The electrophysical and electrochemical properties of living tissue. Ann NY Acad Sci 1974; 238:228-36. 97. Fukada E, Yasuda I. On the piezoelectric effect on bone. J Phys Soc Japan 1957;12:1158-62. 98. Fukada, E Piezoelectric properties of organic polymers. Ann. N.Y. Acad. Sci. 238:7. 1974.

  • 30

    99. Fukuda, E & Yasuda, I. On the piezoelectric effect of bone. J. Physiol. Soc. Japan 10:1158-1162; 1957. 100. Gensler. W Bioelectric potentials and their relation to growth in higher plants. Ann. N.Y. Acad. Sci. 238:280. 1974. 101. Gerling JA, Sinclair P, Roa R The effect of pulsating electromagnetic fields on condylar growth in guinea pigs. Am J Orthod 1985; 87:211-23. 102. Goh JC, Bose K, Kang YK, Nugroho B. Effects of electrical stimulation on the bimechanical properties of fracture healing in rabbits. Clin Orthop 1988;233:268-73. 103. Goodman, R & Henderson, A Exposure of salivary glands to low-frequency electromagnetic fields alters polypeptide synthesis. Proc. Nat. Acad. Sci. U.S.A. 85:3928-3932; 1988. 104. Goodman, R & Henderson, A. S. Sine waves enhance cellular transcription. Bioelectromagnetics 7:23-29; 1986. 105. Graber TM. Extrinsic control factors influencing craniofacial growth. In McNamara JA (ed) Control Mechanisms in Craniofacial Growth, 1975: 75-100. 106. Hamblen, DL. Scientific basis of present day fracture treatment. J. Roy. Col. Surg. of Ed. 24:340-351; 1979. 107. Harrington, DB., Meyer. R & Klein, RM.: Effects of small amounts of electric current at Ihe cellular level. Ann. N.Y. Acad. Sci. 238:300, 1974. 108. Harrington, DB.. and Becker. RO.: Electrical stimulation of RNA and protein synthesis in the frog erylhrocyte. Exp. Cell Res. 76: 95. 1973. 109. Hartshorne. On the causes and treatment of pseudoarthroses and especially that form of it sometimes called supernumary joint. Am. J. Med. Sci. 1:143; 1840. 110. Harvold EP, Vargervik K. Morphogenetic response to activator treatment. Am J Orthod 1971;60:478-90. 111. Hass DW. Stimulation of condylar growth in the cat with pulsating electromagnetic currents. Am J Orthod Dentofacial Orthop 1995;108:599-606. 112. Hassler, CR.; Rykicki, E; Diegle, R & Clark, L. Studies of enhanced bone healing via electrical stimuli. Clin. Orthop. Rel. Res. 124:9-19; 1977.

  • 31

    113. Hasting GW, El Messiery MA, Rakowski S. Mechanoelectrical properties of bone. Biomaterials 1981;2:225-33. 114. Heffeman, M. (1996). Comparative effects of microcurrent stimulation on EEG spectrum and correlation dimension. Integrative Physiology & Behavioral Science. 31 (3):202-209. 115. Heffeman, M. (1996b). Measurement of electromagnetic fields in the healing response. Epress, pp 1-6. 116. Heffernan, M. (1995). The effect of a single cranial electrotherapy stimulation on multiple stress measures. The Townsend Letter for Doctors and Patients. 147:60-64. 117. Heffernan, M. Effects of Variable Microcurrent on EEG Spectra and Pain Control, (1996) ISSSEEM. 118. Henry, H. L. & Norman, A. Vitamin D: Metabolism and mechanism of action. In: Favus, M. J (ed.) Primer on Metabolic Bone Disease. Kelseyville, CA: Amer Society of Bone and Mineral Research; 1990:47-52. 119. Hubel, K. A.: The effects of electrical field stimulation and tetrodotoxin on ion transport by isolated rabbit ileum. J. Clin. Invest. 62:1039. 1977. 120. Jacobs JD, Norton LA. Electrical stimulation of osteogenesis in perioodontal defects. Clin Orthop & Rel Res 1977;124:41-52. 121. Jagendorf. A. T & Uribe. E.: ATP formation caused by acid-base transition of spinach chloroplasts. Proc. Natl. Acad. Sci. U.S.A. 55:170. 1966. 122. Jahn, T. L. A possible mechanism for the effect of electrical potential on apatite formation in bone. Clin. Orthop &. Rel. Res. 56:261-273; 1968. 123. Jorgensen TE. The effects of electric current on the healing time of crural fractures. Acta Orthop Scand 1972;43:421-37. 124. Kantommaa T. The effect of increased oxygen tension on the growth of the mandibular condyle. Acta Odontol Scand 1986;44:307-12. 125. Karpf, DB.; Bambino, T; Arnaud, C; Nissenson, R. Molecular determinants of parathyroid hormone receptor function. In: Cohn, DV; Glorieux, F; Martin, T (eds) Calcium Regulation and Bone Metabolism, vol. 10. Amsterdam: Elsevier; 1990:15-23. " 126. Kaziro Y The role of guanosine-5'-triphosphate in polypeptide chain elongation. Biochem. Biophys. Acta 505:95. 1978.

  • 32

    127. Keller. FB & Zamecnik. PD The effects of guanosine diphosphate and triphosphate on the incorporation of labeled amino acids into proteins. J. Biol. Chem. 221:45, 1956. 128. Killiany DM & Johnston LE. The effect of surgical rotation on the subsequent pattern of condylar growth. In: Dixon AD, Sarnat B & Alan R Liss (eds) Normal and Abnormal Bone Growth. Basic and Clinical Research., New York: 439-48. 129. Kohavi D, Pollack S, Brighton C Short-term effect of guided bone regeneration and electrical stimulation on bone growth in a surgically modelled resorbed dog mandibular ridge. Biomater Artif Cells Immobilization Biotechnol 1992;20:131-8. 130. Kopczyck RA, Norton LA, Kohn MW. Bioelectric regeneration of bone in periodontal defects. J Dent Res 1975; 54:914-19. 131. Korostoff. E.: Stress generated potentials in bone: Relationship to plezoelectricity of collagen. J. Biomech. 10:41. 1977. 132. Koski K, Lahdemaki P. Adaptation of the mandible in children with adenoids. Am J Orthod 1975;68:660-65. 133. Koski KK Cranial growth centers, facts or fallacies? Am J Orthod 1968;54:566-83. 134. Krukowski, M.; Simmons, D. J.; Summerfield, A.; Osdoby, P. Charged beads: Generation of bone and giant cells. J. Bone Min. Res. 2:165-171; 1988. 135. Lanyon. L. E.. and Hartman. W.: Strain related electrical potentials recorded in vitro and in viro. Calcif. Tissue Res. 22:315. 1977. 136. Lavine LS, Lustrin I, Shamos MH & Moss M. The influence of electric current on bone regeneration in vivo. Acta Orthop Scan 1971:42:305-314. 137. Lavine LS, Lustrin I, Shamos M, Rinolds R & Liboff A. Electrical enhancement of bone healing Science 1972;75:1118-21. 138. Lavine. L. S.. Lustfin, I & Shamos. M. H.: Treat C~cal Onnopaeocs and related Rescaeca men: of congenital pseudarthrosis of the tibia with direct current. Clin. Orthop. 124:69. 1977. 139. Levy, D. D & Rubin, B. Inducing bone growth in vivo by pulse stimulation. Clin. Orthop & Rel. Res. 88:218-222; 1972. 140. Liboff, AR.; Williams, T.; Strong, D; Wistar, R. Time-varying magnetic fields: Effect on DNA synthesis. Science 223:818-820; 1984.

  • 33

    141. Liss, S. (1996). Neurochemical profiles following electrocranial stimulation. Presented at the Hans Selye Eighth International Conference on Stress. Montreux, Switzerland. 142. Llaurado JG, Sances A Jr & Battocletti J (eds) Biologic and Clinical Effects of Low Frequency Magnetic and Eelectric Fields CC Thomas, Springfield, Ill1974 143. Lubar, J., et al. (1995). EEG spectrum in neurofeedback treatment of attention deficit disorder. J of Psycho-educational Assessment. Special issue, Dec. 144. Luben, RA. Comparison of electromagnetic effects on para-thyroid hormone receptors and beta-adrenergic receptors in bone cells. J. Cell Biol. 109:172a; 1989. 145. Luben, RA & Cain, C. Use of hormone receptor activities to investigate the membrane effects of low energy electromagnetic fields. In: Adey, WR.; Lawrence, AF (eds) Nonlinear Electrodynamics in Biological Systems. New York: Plenum Press; 1984:23-34. 146. Luben, RA.; Cain, CD.; Chen, MC.; Rosen, D & Adey, WR. Inhibition of parathyroid hormone actions on bone cells in culture by induced low energy electromagnetic fields. Proc. Nat. Acad. Sci. U.S.A. 79:4180-4184; 1982. 147. Luben, RA.; Cobh, D. V. Effects of parathormone and ealci-tonin on citrate and hyaluronate metabolism in cultured bone. Endocrinology 98:413-419; 1976. 148. Luben, RA.; Huynh, D.; Weinshank, R. L.; Smith, L. E. Molecular cloning of candidate sequences for the mouse osteo-blast parathyroid hormone receptor. In: Cohn, DV; Glorieux, F; Martin, T(eds.) Calcium Regulation and Bone Metabolism, vol. 10. Amsterdam: Elsevier; 1990:39-44. 149. Luben, RA.; Wong, G. L.; Cohn, D. V. Biochemical characterization with parathormone and caicitonin ofisolated bone cells: Provisional identification of osteoclasts and osteoblasts. Endocrinology 99:526-534; 1976. 150. Lundin A & Thore A. Analytical information obtainable by evaluation of the tlme course of firefly biolumincscence in the assay of ATP. Anal. Biochem. 66:47. 1975. 151. Luben, RA. (1991). Effects of low-energy electromagnetic fields (pulsed and dc) on membrane signal transduction processes in biological systems. Health Physics. 61(1): 15-28.

  • 34

    152. Luben, RA., Effects of low-energy electromagnetic fields(pulsed and dc)on membrane signal transduction processes in biological systems.(1991) Health Physics, Vol 61, No.1, pgs 15-28. 153. Lundin A & Thore A Analytical information obtainable by evaluation of the tlme course of firefly biolumincscence in the assay of ATP. Anal. Biochem. 66:47. 1975. 154. Manley Tehan, L, Microcurrent Therapy: Universal Treatment Techniques and Applications. Corona, CA: Manley & Associates; 1994 155. Marsland, TP. Biophysical Studies of Pulsed Electromagnetic Field Interaction with Biological Systems. London: Plenum Press; NATO ASI Series 97; 1985:547-595. 156. Martin, R. B & Gutman, W. The effect of electric fields on osteoporosis of disuse. Calc. Tiss. Res. 25:23-27; 1978. 157. Masureik C, Erikson C. Preliminary clinical evaluation of the effect of small electrical currents on the healing of jaw fractures. Clin Orthop & Rel Res 1977;124:84-91. 158. McClanahan, B. J.; Phillips, R. D. The influence of electric field exposure on bone growth and fracture repair in rats. Bioelectromagnetics 4:11-19; 1983. 159. McComb, RB; Bowers, GN Jr & Posen, S. Alkaline Phosphatase. New York: Plenum Press; 1979. 160. McNamara JA Jr., Carlson DS. Quantitative analysis of temporomandibular joint adaptations to protrusive function. Am J Orthod 1979;76:593-611. 161. Mercola, JM & Kirsch, D. The Basis for Microcurrent Electrical Therapy in Conventional Medical Practice, J of Advancement in Medicine, 1995;8(2): 83-97 162. Mitchell. P Chemiosmotic coupling in oxidative and photosynthetic phosphorylation. Biol. Rev. 41:445 1966. 163. Mitchell. P Vectorial chemistry and the molecular mechanism of chemiosmotic coupling: Power transmission by proticity. Biochem. Soc. Trans. 4:400. 1976. 164. Morgareidge, KR Chipman, MR, Microcurrent Therapy, Physical Therapy Today, Spring 1990:50-53

  • 35

    165. Nair, I.; Morgan, G & Florig, H. Biological effects of Power Frequency Electric and Magnetic Fields. Washington, DC: U.S. Government Printing Office; Office of Technology Assessment, Document OTA-BP-E-53; 1989. 166. Nannmark U, Buch F & Albrektsson T. Influence of direct currents on bone vascular supply. Scand J Plast Reconstr Surg Hand Surg 1988; 22 (2) :113-5. 167. Nessler, J.P & Mass, D Direct current electrical stimulation of tendon healing in vitro. Clinical Orthopedics 1985; 217:303. 168. Neuman, W; Ramp, W. The concept of a bone membrane: Source implication. In: Nichols, G; Wasserman, R (eds.) Cellular Mechanisms for Calcium Transfer and Homeostasis. New York: Academic Press; 1971:197-199. 169. Nordenstrm BEWElectric Man Discover April 1986: 1-11 170. Nordenstrm BEW Biologically Closed Electric Circuits: Clinical, Experimental and Theoretical Evidence for an Additional Circulatory System Nordic Medical PublicationsSweden 1983 171. Norton LA, Hanley K & Twrkeitcz J. Bioelectric perturbations of bone. Angle Orthod 1984;54:73-87. 172. Norton LA, Rodan GA & Bourret LA. Epiphyseal cartilage cAMP changes produced by electrical and mechanical perturbations. Clin Orthop & Rel Res 1977;124:59-68. 173. Norton LA. Effects of a pulsed electromagnetic field on mixed chondroblastic tissue culture. Clin Orthop & Rel Res 1982;167:280-9. 174. Norton LA. In vivo bone growth in a controlled electric field. Ann NY Acad Sci 1975;238:466-77. 175. Norton, L. A.; Rodan, G. A. & Bourret, L Epiphyseal cartilage cAMP changes produced by electrical and mechanical perturbations. Clin. Orthop & Rel. Res. 124:59-68; 1977. 176. OConnor BT, Charlton H, Currey J, Kirby DR &Woods C. Effects of electric current on bone in vivo. Nature 1969;222:162-67. 177. O'Malley, B. The steroid hormone receptor superfamily: More excitement predicted for the future. Mol. Endocrinol. 4:363-369; 1990. 178. Otter J, Johnson GS, Pastan IH. Regulation of cell growth cyclic adenosine 3, 5 monophosphate: effect of the cell density and agents which alter growth on cAMP levels in fibroblasts. J Biol Chem 1972;247:7082.

  • 36

    179. Oxender. D. L & Christensen. H Distinct mediating systems for the transport of neutral amino acids by the Ehrlich cell. J. Biol. Chem. 238:3686, 1963. 180. Petrovic A. Mechanism and regulation of condylar growth. Acta Morphol Neerl Scand 1972;10:25-34. 181. Pilla, A. & Margules. G Dynamic interfacial electrochemical phenomena at living cell membranes: Application to the toad urinary bladder membrane system. J. Electrochem. Soc. 124:1697, 1977. 182. Pilla, A. Electrochemical information transfer at cell surfaces and junctions - application to the study and manipulation of cell regulation. In: Keyzer, H. & Gutman, F (eds) Bioelectrochemistry. New York: Plenum Publishing; 1980:353-396. 183. Pinkard JS. The effects of Electric Current on Condylar Cartilage and Bone Growth. Master Thesis, Case Western Reserve University, 1984. 184. Presman AS Electromagnetic Fields and Life Translated by F Sinclair, edited by FE Brown Jr Plenum Press 1970 185. Raisz, LG. Bone metabolism and calcium regulation. Metabolic Bone Dis. 1:1-48; 1977. 186. Rappaport, M. S.; Stern, P. H. Parathyroid hormone and calcitonin modify inositol phospholipid metabolism in fetal rat limb bones. J. Bone Min. Res. 1:173-179; 1986. 187. Rodan GA, Barret L, Harvey A & Mens T. Cyclic AMP and cyclic GMP: Mediators of the mechanical effects on bone remodeling. Science 1975; 189:467-9. 188. Rodan GA, Bourret LA & Norton LA. DNA synthesis in cartilage cells is stimulated by oscillating electric fields. Science 1978;199:690-2. 189. Rodan, GA.; Bourret, L. A.; Norton, L. A. DNA synthesis in cartilage cells is stimulated by oscillating electric fields. Science 199:690--692; 1978. 190. Rodan, GA.; Martin, T. J. Role of osteoblasts in the hormonal regulation of bone resorption--a hypothesis. Calcif. Tiss. Intl. 33:349-351; 1981. 191. Rodan, S. B.; Wesolowski, G.; Rodan, G. A. Cional differences in prostaglandin synthesis among osteosarcoma cell lines. J. Bone Min. Res. 1:213-220; 1986.

  • 37

    192. Romero-Sierra I & Tanner. JA.: Biological effects of nonionizing radiation: An outline of fundamenlal laws. Ann. N.Y. Acad. Sci. 238:263, 1974. 193. Rowley, BA; McKenna, J & Wolcott, L; The use of Low Level Electric Current for the Enhancement of Tissue Healing. ISA BM. 1974; 74322: 111-114 194. Rowley BA, McKenna, J; Chase, G & Wolcott, L The influence of electrical current on an infecting microorganism in wounds. Ann. N.Y. Acad. Sci. 238:543, 1974. 195. Sansen W & De Dijcker, F.: The four-point probe technique to measure bio-impedances. Electromyogr. Clin. Neurophysiol. 16:509. 1976. 196. Savitz, D. A.; Calle, E. Leukemia and occupational exposure to electromagnetic fields: Review of epidemiological surveys. J. Med. 29:47-51; 1987. 197. Schlessinger, J. The epidermal growth factor receptor as a mul-tifunctional allosteric protein. Biochem. 27:3119-3123; 1986. 198. Schmukler, R.; Pilla, A. A. A transient impedance approach to nonfaradaic electrochemical kinetics at living cell membranes. J. Electrochem. Soc. 129:526-528; 1982. 199. Selye, HThe Stress of Life. New York: Van Nostrand-Reinhold; 1975 200. Shafer DM, Rogerson K, Norton L, Bennett J. The effect of electrical perturbation on osseointegration of titanium dental implants: a preliminary study. J Oral Maxillofac Surg 1995;53:1063-8. 201. Shamos MH. & Layine LS.: Piezoelectricity as a fundamental property of biological tissues. Nature 213:267, 1967. 202. Shapiro E, Roeber FW, Klempner LS. Orthodontic movement using pulsating force-induced piezoelectricity. Am J Orthod 1975;76:251-254. 203. Sibley, D. R.; Benovic, J; Caron, M; Lefkowitz, R. Phosphorylation of cell surface receptors: A mechanism for regulating signal transduction pathways. Endocrine Rev. 9: 38-56; 1988. 204. Spadaro JA, Albanese SA, Chase S. Bone formation near direct current electrodes with and without motion. J Orthop Res 1992;10,5:729-38. 205. Spafaro. J. A.: Electrically stimulated bone growth in animals and man. Review of the literature. Clin Orthop & Rel Res. 122:325, 1977.

  • 38

    206. Sparado JA & Becker RO. Function of implanted cathodes in elecrode-induced bone growth. Med Biol Eng Comput 1979;17:769-75. 207. Sparado JA. Electrically stimulated bone growth in animals and man. Clin Orthop & Rel Res 1977;122:325-32. 208. Stan. S, Muller, J; Sansen. W. & Dewaele. P.: Effect of direct current on the healing of fractures. In Burney. F.. Herbst, E. & Hinsenkamp. M. (eds): Electric Stimulation of Bone Growth and Repair. Berlin. Heidelberg. New York, Springer-Verlag. 1978, pp. 47-53. 209. Stark TM, Sinclair PM. Effect of pulsed electromagnetic fields on orthodontic tooth movement. Am J Orthod 1987;91:91-103. 210. Starlanyl, D Fibromyalgia & Chronic Myofascial Pain Syndrome: A Survival Manual. Oakland, CA: New Harbinger Publications, Inc.; 1996 211. Stefan A, Sanses W, Milier JC. Experimental study on electrical impedance on bone and the effect of direct on the healing of fractures. Clin Orthop a Rel Res 1976;120:264-67. 212. Steiner M & Ramp WK. Electrical stimulation of bone and its applications for endosseous dental implantation. J Oral Implantol 1990;16:20-7. 213. Stryer, L & Bourne, HR . G proteins: A family of signal transducers. Ann. Rev. Cell Biol. 2:391-420; 1986. 214. Stutzman J, Petrovic. Intrinsic regulation of the condylar cartilage growth rate. Europ J Orthod 1979;1:41-54. 215. Szago G, Illes T. Experimental stimulation of osteogenesis induced by bone matrix. Orthopedics 1991;14,1:63-7. 216. Tabrah, F.; Hoffmeier, M.; Gilbert, F.; Batkin, S.; Bassett, C. A. Bone density changes in osteoporosis-prone women exposed to pulsed electromagnetic fields (PEMFs). J. Bone Min. Res. 5:437-442; 1990. 217. Tam, CS.; Heersche, J; Murray, T & Parsons, J. Parathyroid hormone stimulates the bone apposition rate independently of its resorptive action. Endocrinology 110:506-512; 1982. 218. Travell, JG & Simons, DG Myofascial Pain and Dysfunction:The Trigger Point Manual, Vol. 1: The Upper Body. Baltimore, MD: Williams & Wilkins; 1983 219. United States Environmental Protection Agency. Evaluation of the potential carcinogenicity of Electromagnetic Fields. July 1991, Volume 61, Number I

  • 39

    220. Van Linborgh J. A new view on the control of the morphogenesis of the skull. Acta Morphol Neerl Scand 1970;8:143-60. 221. Vingerling PA, Van der Kuij P, De Groot K & Sillevis PAE. Electromagnetic reduction of resorption rete of extrxction wounds. In: Brighton CT, Black J, Pollack SR (eds) Electrical Properties of Bone and Cartilage. New York: Grune & Straton, 1979:341-6. 222. Wahlstrom, O. Stimulation of fracture healing with electromagnetic fields of extremely low frequency. Clin. Orthop 186:293-298; 1984. 223. Watson, J. The electrical stimulation of bone healing. Proc. IEEE 67:1339-1351; 1979. 224. Weislander L, Lagerstrom L. The effect pf activator on Class II malocclusions. Am J Orthod 1979;75:20-26. 225. West, B.J. (1990). Fractal Physiology and Chaos in Medicine. World Scientific, New Jersey. 226. Witt. H. T.. Schlodder, E & Graber. P Membrane-bound ATP synthesis generated by an external electrical field. FEBS Left. 69:272, 1976. 227. Wolcott. L. E.. Wheeler, P, Hardwicke. H & RowIey. B Accelerated healing of skin ulcers by electrotherapy: Prelimlnary clinical results. South. Med. J. 62:795. 1969. 228. Wolff, J. Studies of Bone Transformation. Berlin: Publisher unknown; 1892. 229. Wu. K. T., Go, N.. Dennis, C.. Enquist, I. & Sawyer P. N. Effects of Electric Currents and interfacial potentials on wound healing. J Surg. Res. 7:122. 1967. 230. Yamaguchi, D. T.; Hahn, T; Lida-Klein, A; Kleeman, C & Muallemm, S. Parathyroid hormone activated calciun channels in an osteoblast-like clonal osteosarcoma cell line. J. Biol. Chem. 262:7711-7718; 1987. 231. Yarden, Y.; Ullrich, A. Growth factor receptor tyrosine kinases Ann. Rev. Biochem. 57:443-478; 1988 232. Yasuda I. Electrical callus and callus formation by electret. Clin Orthop & Rel Res 1977;124:53-56. 233. Zengo AN, Bassett CA, Proutzos G, Pawluk R, Pilla A. In vivo effects of direct current in the mandible. J Dent Res 1976; 58:383-90.

  • 40